WO2015024669A1 - Vaccin combiné - Google Patents

Vaccin combiné Download PDF

Info

Publication number
WO2015024669A1
WO2015024669A1 PCT/EP2014/002302 EP2014002302W WO2015024669A1 WO 2015024669 A1 WO2015024669 A1 WO 2015024669A1 EP 2014002302 W EP2014002302 W EP 2014002302W WO 2015024669 A1 WO2015024669 A1 WO 2015024669A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
rna
protein
combination vaccine
virus
Prior art date
Application number
PCT/EP2014/002302
Other languages
English (en)
Inventor
Karl-Josef Kallen
Thomas Kramps
Margit SCHNEE
Daniel Voss
Original Assignee
Curevac Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curevac Gmbh filed Critical Curevac Gmbh
Priority to CA2915730A priority Critical patent/CA2915730A1/fr
Priority to AU2014310935A priority patent/AU2014310935B2/en
Priority to EP14761282.4A priority patent/EP3035959A1/fr
Priority to CN201480044258.XA priority patent/CN105473157A/zh
Publication of WO2015024669A1 publication Critical patent/WO2015024669A1/fr
Priority to US15/048,561 priority patent/US10588959B2/en
Priority to US16/781,781 priority patent/US11266735B2/en
Priority to US17/590,173 priority patent/US20220152193A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/155Paramyxoviridae, e.g. parainfluenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, e.g. influenza virus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18534Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present invention relates to a vaccine, especially a combination vaccine providing at leasta first and a second antigenic function, wherein the antigenic functions are encoded by at least one mRNA encoding at least one or more proteins or fragments, variants or derivatives of proteins awarding antigenic function, wherein the first antigenic function being a Fusion (F) protein or a fragment, variant or derivative of a Fusion (F) protein derived from the virus family Paramyxoviridae and the second antigenic function being an Hemagglutinin (HA) protein or a fragment, variant or derivative of an Hemagglutinin (HA) protein derived from the virus family Orthomyxoviridae.
  • the present invention is directed to a kit or kit of parts comprising the components of said combination vaccine and to said combination vaccine for use in a method of prophylactic or therapeutic treatment of diseases, particularly in the prevention or treatment of infectious diseases like RSV and influenza.
  • Respiratory diseases caused by viruses or bacteria are a major health and economic burden worldwide.
  • most prominent viral pathogens are respiratory syncytial virus (RSV), parainfluenza viruses 1 -3 (PIV), and influenza A and B viruses, which are responsible for the majority of lower respiratory tract infections resulting in a significant rate of hospitalizations particularly of young children less than 3 years of age (Forster, J. et al., 2004. Prospective population-based study of viral lower respiratory tract infections in children under 3 years of age (the PRI.DE study). European Journal of Pediatrics, 1 63(12), S.709-716.).
  • RSV which belongs to the virus family of Paramyxoviridae, is one of the most contagious pathogens and makes a substantial contribution to severe respiratory tract infections in infants, the elderly and immunocompromised patients.
  • PIV parainfluenza viruses
  • the subtypes 1 and 2 of PIV are the principal causes of croup, whereas subtype 3 causes more severe lower respiratory tract illness with RSV-like symptoms including pneumonia and bronchiolitis.
  • Paramyxoviruses are also responsible for a range of diseases in other animal species, for example canine distemper virus (dogs), phocine distemper virus (seals), cetacean morbillivirus (dolphins and porpoises), Newcastle disease virus (birds), and rinderpest virus (cattle).
  • HeV Henipavirus
  • Nipah virus Nipah virus
  • Paramyxoviridae typically do express a so called Fusion (F) protein which projects from the virus envelope surface and mediates cell entry by inducing a fusion process between the virus and the cell to be infected.
  • F Fusion
  • Influenza viruses belong to the virus family Orthomyxoviridae and pose a high risk especially for infants, children and the elderly. Influenza viruses possess a segmented, negative-stranded RNA genome and are divided into three main types A, B, and C, of which type A is the most prominent one in humans. Influenza A viruses can be further subdivided based on different forms of the two surface glycoproteins Hemagglutinin (HA) and Neuraminidase (NA).
  • HA Hemagglutinin
  • NA Neuraminidase
  • the impact of seasonal influenza characteristically a febrile disease with respiratory syndromes, has been estimated at 25-50 million cases per year worldwide. Due to the possibility of re-assortment of genetic material new variants of influenza viruses can emerge sporadically and spread worldwide (pandemic). Such re-assortment occurs most readily in pigs ("mixing vessels") resulting e.g. in the genesis of the swine-origin H1 N1 in 2009 (“swine flu”).
  • influenza vaccines are subunit, inactivated split or whole virion vaccines propagated in cell culture or chicken eggs which are not recommended for infants and only limited recommended for pregnant women.
  • a humanised monoclonal antibody against the viral surface F protein is the only prophylactic product on the market which is recommended for infants considered at high risk including pre-term infants and infants with chronic lung disease (The IMpact- RSV Study Group. 1998. Palivizumab, a Humanized Respiratory Syncytial Virus Monoclonal Antibody, Reduces Hospitalization From Respiratory Syncytial Virus Infection in High-risk Infants. Pediatrics, 102(3), S.531 -537., Tablan et a/. 2003. Guidelines for preventing health- care-associated pneumonia, 2003: recommendations of CDC and the Healthcare Infection Control Practices Advisory Committee. MMWR.
  • Recommendations and Reports Morbidity and Mortality Weekly Report. Recommendations and Reports / Centers for Disease Control, 53(RR-3), S.1 -36.).
  • Recent studies with animal models demonstrated that sufficient amounts of neutralising antibodies targeting RSV F protein limit viral replication leading to a less severe course of disease (Singh, S.R. eta/., 2007.
  • WO 201 1/030218 discloses immunogenic compositions comprising viral (RSV and influenza) and bacterial (pneumococcus) immunogens
  • WO 00/35481 discloses combinations of RSV F, G and matrix proteins with a non-virulent influenza virus preparation
  • WO 2010/149743 discloses combinations of F proteins derived from human metapneumovirus, parainfluenza virus and RSV.
  • Talaat et a/. Talaat, A.M. et a/. 2001 .
  • a combination vaccine confers full protection against co-infections with influenza, herpes simplex and respiratory syncytial viruses.
  • Vaccine, 20(3-4), S.538-544) disclose a combination of DNA plasmid-driven vaccines against RSV, Herpes simplex virus (HSV) and Influenza A. Such a strategy, however, still requires administration of DNA based vectors.
  • a further drawback, however, is the unknown compatibility between different co-administered novel vaccines e.g. by antigen competition.
  • the object of the invention to provide a further vaccine or possibly even an improved vaccine.
  • a (combination) vaccine against respiratory diseases caused by viruses of the Paramyxoviridae and/or the Orthomyxoviridae iamW are particularly caused by RSV and/or influenza viruses.
  • a pharmaceutical composition or a kit comprising the (combination) vaccine or the respective components thereof. It is an object to provide a (combination) vaccine for use in a method of treatment of infections caused by viruses of the virus families Paramyxoviridae, e.g. RSV, and/or Orthomyxoviridae, e.g. Influenza virus.
  • viruses of the virus families Paramyxoviridae e.g. RSV
  • Orthomyxoviridae e.g. Influenza virus.
  • RSV respiratory syncytial virus
  • PAV parainfluenza viruses 1 -3
  • Influenza A and B viruses which induce a balanced immune response, i.e. a humoral and a cellular immune response.
  • Genome of SV RSV has 10 genes encoding 1 1 proteins— there are 2 open reading frames of M2. NS1 and NS2 inhibit type I interferon activity. N encodes nucleocapsid protein that associates with the genomic RNA forming the nucleocapsid. M encodes the Matrix protein required for viral assembly. SH, G and F form the viral coat. The "G" protein is a surface protein that is heavily glycosylated.
  • the "F” protein is another important surface protein; F mediates fusion, allowing entry of the virus into the cell cytoplasm and also allowing the formation of syncytia.
  • the "F” protein is homologous in both subtypes of RSV; antibodies directed against the "F” protein are neutralizing.
  • the "G” protein differs considerably between the two subtypes.
  • M2 is the second matrix protein also required for transcription, it encodes M2-1 (elongation factor) and M2-2 (transcription regulation), M2 contains CD8 epitopes.
  • L encodes the RNA polymerase.
  • the phosphoprotein P is a cofactor for L.
  • Genome of Influenza Despite of all variations, the viral particles of all influenza viruses are similar in composition. These are made of a viral envelope containing two main types of glycoproteins, wrapped around a central core. The central core contains the viral RNA genome and other viral proteins that package and protect this RNA. Unusually for a virus, its genome is not a single piece of nucleic acid; instead, it contains seven or eight pieces of segmented negative-sense RNA, each piece of RNA containing either one or two genes, which code for a gene product (protein).
  • influenza A genome contains 1 1 genes on eight pieces of RNA, encoding 1 1 proteins: hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), M1, M2, NS1 , NS2 (NEP: nuclear export protein), PA, PB1 (polymerase basic 1 ), PB1 -F2 and PB2.
  • Hemagglutinin (HA) and neuraminidase (NA) are the two large glycoproteins on the outside of the viral particles.
  • HA is a lectin that mediates binding of the virus to target cells and entry of the viral genome into the target cell, while NA is involved in the release of progeny virus from infected cells, by cleaving sugars that bind the mature viral particles. Furthermore, they are antigens to which antibodies can be raised. Influenza A viruses are classified into subtypes based on antibody responses to HA and NA. These different types of HA and NA form the basis of the H and N distinctions in, for example, H5N1 . There are 1 6 H and 9 N subtypes known, but only H 1 , 2 and 3, and N 1 and 2 are commonly found in humans.
  • Adaptive immune response is typically understood to be antigen-specific. Antigen specificity allows for the generation of responses that are tailored to specific antigens, pathogens or pathogen-infected cells. The ability to mount these tailored responses is maintained in the body by "memory cells". Should a pathogen infect the body more than once, these specific memory cells are used to quickly eliminate it.
  • the first step of an adaptive immune response is the activation of naive antigen- specific T cells or different immune cells able to induce an antigen-specific immune response by antigen-presenting cells. This occurs in the lymphoid tissues and organs through which naive T cells are constantly passing.
  • Dendritic cells that can serve as antigen-presenting cells are inter alia dendritic cells, macrophages, and B cells. Each of these cells has a distinct function in eliciting immune responses.
  • Dendritic cells take up antigens by phagocytosis and macropinocytosis and are stimulated by contact with e.g. a foreign antigen to migrate to the local lymphoid tissue, where they differentiate into mature dendritic cells.
  • Macrophages ingest particulate antigens such as bacteria and are induced by infectious agents or other appropriate stimuli to express MHC molecules.
  • the unique ability of B cells to bind and internalize soluble protein antigens via their receptors may also be important to induce T cells.
  • T cells which induces their proliferation and differentiation into armed effector T cells.
  • the most important function of effector T cells is the killing of infected cells by CD8+ cytotoxic T cells and the activation of macrophages by Th1 cells which together make up cell-mediated immunity, and the activation of B cells by both Th2 and Th1 cells to produce different classes of antibody, thus driving the humoral immune response.
  • T cells recognize an antigen by their T cell receptors which do not recognize and bind antigen directly, but instead recognize short peptide fragments e.g. of pathogen-derived protein antigens, which are bound to MHC molecules on the surfaces of other cells.
  • the adaptive immune system is composed of highly specialized, systemic cells and processes that eliminate or prevent pathogenic growth.
  • the adaptive immune response provides the vertebrate immune system with the ability to recognize and remember specific pathogens (to generate immunity), and to mount stronger attacks each time the pathogen is encountered.
  • the system is highly adaptable because of somatic hypermutation (a process of increased frequency of somatic mutations), and V(D)J recombination (an irreversible genetic recombination of antigen receptor gene segments). This mechanism allows a small number of genes to generate a vast number of different antigen receptors, which are then uniquely expressed on each individual lymphocyte.
  • Immune network theory is a theory of how the adaptive immune system works, that is based on interactions between the variable regions of the receptors of T cells, B cells and of molecules made by T cells and B cells that have variable regions.
  • Adjuvant / adjuvant component An adjuvant or an adjuvant component in the broadest sense is typically a (e.g. pharmacological or immunological) agent or composition that may modify, e.g. enhance, the efficacy of other agents, such as a drug or vaccine.
  • a (e.g. pharmacological or immunological) agent or composition that may modify, e.g. enhance, the efficacy of other agents, such as a drug or vaccine.
  • the term refers in the context of the invention to a compound or composition that serves as a carrier or auxiliary substance for immunogens and/or other pharmaceutically active compounds. It is to be interpreted in a broad sense and refers to a broad spectrum of substances that are able to increase the immunogenicity of antigens incorporated into or coadministered with an adjuvant in question.
  • an adjuvant will preferably enhance the specific immunogenic effect of the active agents of the present invention.
  • adjuvant or “adjuvant component” has the same meaning and can be used mutually.
  • Adjuvants may be divided, e.g., into immuno potentiators, antigenic delivery systems or even combinations thereof.
  • adjuvant is typically understood not to comprise agents which confer immunity by themselves.
  • An adjuvant assists the immune system unspecifically to enhance the antigen-specific immune response by e.g. promoting presentation of an antigen to the immune system or induction of an unspecific innate immune response.
  • an adjuvant may preferably e.g. modulate the antigen-specific immune response by e.g. shifting the dominating Th2-based antigen specific response to a more Thl -based antigen specific response or vice versa. Accordingly, an adjuvant may favourably modulate cytokine expression/secretion, antigen presentation, type of immune response etc.
  • the term "antigen" refers typically to a substance which may be recognized by the immune system and may be capable of triggering an antigen-specific immune response, e.g. by formation of antibodies or antigen- specific T-cells as part of an adaptive immune response.
  • An antigen may be a protein or peptide.
  • the first step of an adaptive immune response is the activation of naive antigen-specific T cells by antigen-presenting cells. This occurs in the lymphoid tissues and organs through which naive T cells are constantly passing.
  • the three cell types that can serve as antigen-presenting cells are dendritic cells, macrophages, and B cells. Each of these cells has a distinct function in eliciting immune responses.
  • Tissue dendritic cells take up antigens by phagocytosis and macropinocytosis and are stimulated by infection to migrate to the local lymphoid tissue, where they differentiate into mature dendritic cells. Macrophages ingest particulate antigens such as bacteria and are induced by infectious agents to express MHC class II molecules. The unique ability of B cells to bind and internalize soluble protein antigens via their receptors may be important to induce T cells. By presenting the antigen on MHC molecules leads to activation of T cells which induces their proliferation and differentiation into armed effector T cells.
  • effector T cells The most important function of effector T cells is the killing of infected cells by CD8 + cytotoxic T cells and the activation of macrophages by TH1 cells which together make up cell-mediated immunity, and the activation of B cells by both TH2 and TH1 cells to produce different classes of antibody, thus driving the humoral immune response.
  • T cells recognize an antigen by their T cell receptors which does not recognize and bind antigen directly, but instead recognize short peptide fragments e.g. of pathogens' protein antigens, which are bound to MHC molecules on the surfaces of other cells.
  • T cells fall into two major classes that have different effector functions. The two classes are distinguished by the expression of the cell-surface proteins CD4 and CD8.
  • T cells differ in the class of MHC molecule that they recognize.
  • MHC molecules There are two classes of MHC molecules - MHC class I and MHC class II molecules - which differ in their structure and expression pattern on tissues of the body.
  • CD4 + T cells bind to a MHC class II molecule and CD8 + T cells to a MHC class I molecule.
  • MHC class I and MHC class II molecules have distinct distributions among cells that reflect the different effector functions of the T cells that recognize them.
  • MHC class I molecules present peptides of cytosolic and nuclear origin e.g. from pathogens, commonly viruses, to CD8 + T cells, which differentiate into cytotoxic T cells that are specialized to kill any cell that they specifically recognize.
  • MHC class I molecules Almost all cells express MHC class I molecules, although the level of constitutive expression varies from one cell type to the next. But not only pathogenic peptides from viruses are presented by MHC class I molecules, also self-antigens like tumour antigens are presented by them. MHC class I molecules bind peptides from proteins degraded in the cytosol and transported in the endoplasmic reticulum. The CD8 + T cells that recognize MHC class hpeptide complexes at the surface of infected cells are specialized to kill any cells displaying foreign peptides and so rid the body of cells infected with viruses and other cytosolic pathogens.
  • CD4 + T cells CD4 + helper T cells
  • MHC class II molecules are normally found on B lymphocytes, dendritic cells, and macrophages, cells that participate in immune responses, but not on other tissue cells. Macrophages, for example, are activated to kill the intravesicular pathogens they harbour, and B cells to secrete immunoglobulins against foreign molecules. MHC class II molecules are prevented from binding to peptides in the endoplasmic reticulum and thus MHC class II molecules bind peptides from proteins which are degraded in endosomes.
  • TH1 cells can capture peptides from pathogens that have entered the vesicular system of macrophages, or from antigens internalized by immature dendritic cells or the immunoglobulin receptors of B cells.
  • Pathogens that accumulate in large numbers inside macrophage and dendritic cell vesicles tend to stimulate the differentiation of TH1 cells, whereas extracellular antigens tend to stimulate the production of TH2 cells.
  • TH1 cells activate the microbicidal properties of macrophages and induce B cells to make IgG antibodies that are very effective of opsonising extracellular pathogens for ingestion by phagocytic cells
  • TH2 cells initiate the humoral response by activating naive B cells to secrete IgM, and induce the production of weakly opsonising antibodies such as IgGI and lgG3 (mouse) and lgG2 and lgG4 (human) as well as IgA and IgE (mouse and human).
  • a vaccine is typically understood to be a prophylactic or therapeutic material providing at least one antigen or antigenic function.
  • the antigen or antigenic function may stimulate the body's adaptive immune system to provide an adaptive immune response.
  • Antibacterial agent An antibacterial agent is typically a substance that may be effective against bacteria. The antibacterial agent may for example directly kill bacteria, reduce bacterial growth, and/or inhibit bacterial propagation and spreading. Examples for antibacterial agents are given further below.
  • Antiviral agent An antiviral agent is typically a substance that may be effective against viruses. The antiviral agent may for example directly inactivate viruses, reduce viral replication, and/or inhibit viral propagation and spreading. Examples for antibacterial agents are given further below.
  • Antigenic function may for example be an immunogen.
  • Antigenic functions in the context of the present invention also encompass mediators, i.e. nucleic acids which do show an antigenic function in vivo if they code for antigenic proteins/peptides.
  • mediators i.e. nucleic acids which do show an antigenic function in vivo if they code for antigenic proteins/peptides.
  • Such carriers having antigenic function as understood in the context of the inventions may be expressed by the nucleic acid in vivo which in turn leads to the presence of proteins or peptides that may act as an immunogen.
  • an antigenic function is typically a component that can lead directly (direct antigenic functionality / directly acting antigenic function) or indirectly (indirect antigenic functionality / indirectly acting antigenic function) to the presence of an antigen within an organism when introduced into this organism.
  • direct antigenic functionality typically means that the antigenic function is, e.g., a protein or peptide (or a killed bacterium, virus or the like) that is administered to an organism and induces an adaptive immune response, mostly without being modified by e.g. translation or the like.
  • indirect antigenic functionality typically means in this context that the "antigenic function” is, e.g., a nucleic acid sequence that is taken up by the target organism and translated within the organism into a peptide or protein. This peptide or protein then functions as an immunogen and induces an adaptive immune response.
  • an "antigenic function" is understood to be a preform or precursor of an immunogen.
  • an "antigenic function" can be understood to be an immunogen itself.
  • an antigenic function may in particular be a Fusion (F) protein of the virus family Paramyxoviridae and (e.g. artificial) functional variants or fragments thereof as well as (preferably immunogenic) fragments of said Fusion (F) protein and respective variants; as well as corresponding nucleic acids encoding any of these, i.e. Fusion (F) proteins of the virus family Paramyxoviridae, variants thereof as well as fragments of said Fusion (F) protein and respective variants.
  • an antigenic function may also in particular be a Hemagglutinin (HA) protein of the virus family Orthomyxoviridae and (e.g. artificial) variants thereof as well as (preferably immunogenic) fragments of said Hemagglutinin (HA) protein and respective variants; as well as corresponding nucleic acids encoding any of these, i.e. Hemagglutinin (HA) proteins of the virus family Orthomyxoviridae, variants thereof as well as fragments of said Hemagglutinin (HA) protein and respective variants.
  • HA and F protein may for example be identified in established databases such as the UniProt database or the Protein database provided by the National Center for Biotechnology (NCBI, US).
  • Hemagglutinin (HA) proteins of the virus family Orthomyxoviridae and (e.g. artificial) variants thereof may for instance likewise be identified in databases such as the UniProt database or the Protein database provided by the National Center for Biotechnology (NCBI, US).
  • Antigenic function preferably represents the immune response elicited by a protein or peptide sequence.
  • the antigenic function or the antigenic potential of the HA and F protein is typically sequence specific and depends on specific epitope sequences within the full-length protein.
  • the antigenis function in terms of the T cell response typically depends on T cell epitopes, which is typically evoked by peptide (fragments) of a length of between 8 and 1 1 amino acids (for presentation by MHC class I molecules), whereas B cell epitopes (for presentation on MHC class II molecules) are typically longer peptides of 1 3-1 7 amino acids in length.
  • the antigenic function(s) may preferably be understood as the immunological potential or immunogenicity (for triggering a T- and B cell response), which is due to the characteristic T and B cell epitopes of the full-length protein, e.g. the HA or F protein.
  • the fragments, variants or derivatives of the full-length protein shall typically retain the same immunological potential as the full-length HA or F proteins to reflect their antigenic function.
  • Antigen-providing RNA in particular an antigen-providing mRNA in the context of the invention may typically be a RNA, having at least one open reading frame that can be translated by a cell or an organism provided with that RNA.
  • the product of this translation is a peptide or protein that may act as an antigen, preferably as an immunogen.
  • the product may also be a fusion protein composed of more than one immunogen, e.g. a fusion protein that consist of two or more epitopes, peptides or proteins derived from the same or different virus-proteins, wherein the epitopes, peptides or proteins may be linked by linker sequences.
  • RNA preferably an mRNA, that typically may have two (bicistronic) or more (multicistronic) open reading frames (ORF).
  • An open reading frame in this context is a sequence of several nucleotide triplets (codons) that can be translated into a peptide or protein. Translation of such RNA yields two (bicistronic) or more (multicistronic) distinct translation products (provided the ORFs are not identical).
  • such RNA may for example comprise an internal ribosomal entry site (IRES) sequence.
  • Fragments or variants of nucleic acids may typically comprise a sequence having a sequence identity with a nucleic acid, or with a protein or peptide, if encoded by the nucleic acid molecule, of at least 5%, 1 0%, 20%, 30%, 40%, 50%, 60%, preferably at least 70%, more preferably at least 80%, equally more preferably at least 85%, even more preferably at least 90% and most preferably at least 95% or even 97%, 98% or 99%, to the entire wild type sequence, either on nucleic acid level or on amino acid level.
  • Carrier / polymeric carrier A carrier in the context of the invention may typically be a compound that facilitates transport and/or complexation of another compound. Said carrier may form a complex with said other compound.
  • a polymeric carrier is a carrier that is formed of a polymer.
  • Cationic component typically refers to a charged molecule, which is positively charged (cation) at a pH value of typically about 1 to 9, preferably of a pH value of or below 9 (e.g. 5 to 9), of or below 8 (e.g. 5 to 8), of or below 7 (e.g. 5 to 7), most preferably at physiological pH values, e.g. about 7.3 to 7.4. Accordingly, a cationic peptide, protein or polymer according to the present invention is positively charged under physiological conditions, particularly under physiological salt conditions of the cell in vivo.
  • a cationic peptide or protein preferably contains a larger number of cationic amino acids, e.g.
  • a 5' cap is typically a modified nucleotide, particularly a guanine nucleotide, added to the 5' end of a RNA-molecule. Preferably, the 5'cap is added using a 5 '-5 '-triphosphate linkage.
  • Cellular immunity/cellular immune response relates typically to the activation of macrophages, natural killer cells (NK), antigen-specific cytotoxic T- lymphocytes, and the release of various cytokines in response to an antigen.
  • cellular immunity is not related to antibodies but to the activation of cells of the immune system.
  • a cellular immune response is characterized e.g.
  • cytotoxic T-lymphocytes that are able to induce apoptosis in body cells displaying epitopes of an antigen on their surface, such as virus-infected cells, cells with intracellular bacteria, and cancer cells displaying tumor antigens; activating macrophages and natural killer cells, enabling them to destroy pathogens; and stimulating cells to secrete a variety of cytokines that influence the function of other cells involved in adaptive immune responses and innate immune responses.
  • a combination vaccine is typically a vaccine that may provide two or more immunogens and/or antigenic functions. The immunogens and/or antigenic functions are provided simultaneously by one composition.
  • Fragments of proteins in the context of the present invention may, typically, comprise a sequence of a protein or peptide as defined herein, which is, with regard to its amino acid sequence (or its encoded nucleic acid molecule), N- terminally and/or C-terminally truncated compared to the amino acid sequence of the original (native) protein (or its encoded nucleic acid molecule). Such truncation may thus occur either on the amino acid level or correspondingly on the nucleic acid level.
  • a sequence identity with respect to such a fragment as defined herein may therefore preferably refer to the entire protein or peptide as defined herein or to the entire (coding) nucleic acid molecule of such a protein or peptide.
  • fragments of nucleic acids in the context of the present invention may comprise a sequence of a nucleic acid as defined herein, which is, with regard to its nucleic acid molecule 5'- and/or 3'- truncated compared to the nucleic acid molecule of the original (native) nucleic acid molecule.
  • a sequence identity with respect to such a fragment as defined herein may therefore preferably refer to the entire nucleic acid as defined herein.
  • Fragments of proteins or peptides in the context of the present invention may furthermore comprise a sequence of a protein or peptide as defined herein, which has a length of for example at least 5 amino acids, preferably a length of at least 6 amino acids, preferably at least 7 amino acids, more preferably at least 8 amino acids, even more preferably at least 9 amino acids; even more preferably at least 10 amino acids; even more preferably at least 1 1 amino acids; even more preferably at least 12 amino acids; even more preferably at least 13 amino acids; even more preferably at least 14 amino acids; even more preferably at least 15 amino acids; even more preferably at least 1 6 amino acids; even more preferably at least 1 7 amino acids; even more preferably at least 18 amino acids; even more preferably at least 19 amino acids; even more preferably at least 20 amino acids; even more preferably at least 25 amino acids; even more preferably at least 30 amino acids; even more preferably at least 35 amino acids; even more preferably at least 50 amino acids; or most preferably at least 100 amino acids.
  • such fragment may have a length of about 6 to about 20 or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 6, 7, 1 1 , or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 13 or more amino acids, e.g. 13, 14, 15, 16, 1 7, 18, 19, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence.
  • These fragments are typically recognized by T-cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e. the fragments are typically not recognized in their native form.
  • Fragments of proteins or peptides may comprise at least one epitope of those proteins or peptides.
  • domains of a protein like the extracellular domain, the intracellular domain or the transmembrane domain and shortened or truncated versions of a protein may be understood to comprise a fragment of a protein. The fragment may be chosen as mentioned from any part of the full length protein or peptide.
  • the fragment of a Fusion (F) protein of the virus family Paramyxoviridae, and/or the fragment of the Hemagglutinin (HA) protein of the virus family Orthomyxoviridae may be selected, independently of each other, from the first, second, third or fourth quarter of the amino acid sequence of said Fusion (F) protein of the virus family Paramyxoviridae and/or the amino acid sequence of said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae, respectively.
  • T cell epitopes or parts of the proteins in the context of the present invention may comprise fragments preferably having a length of about 6 to about 20 or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 1 1 , or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 13 or more amino acids, e.g. 13, 14, 15, 16, 1 7, 18, 19, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence.
  • These fragments are typically recognized by T cells in form of a complex consisting of the peptide fragment and an MHC molecule.
  • B cell epitopes are typically fragments located on the outer surface of (native) protein or peptide antigens as defined herein, preferably having 5 to 15 amino acids, more preferably having 5 to 12 amino acids, even more preferably having 6 to 9 amino acids, which may be recognized by antibodies, i.e. in their native form.
  • Such epitopes of proteins or peptides may furthermore be selected from any of the herein mentioned variants of such proteins or peptides.
  • antigenic determinants can be conformational or discontinuous epitopes which are composed of segments of the proteins or peptides as defined herein that are discontinuous in the amino acid sequence of the proteins or peptides as defined herein but are brought together in the three-dimensional structure or continuous or linear epitopes which are composed of a single polypeptide chain.
  • Variants of proteins may be generated, having an amino acid sequence which differs from the original sequence in one or more mutation(s), such as one or more substituted, inserted and/or deleted amino acid(s). Preferably, these fragments and/or variants have the same biological function or specific activity compared to the full-length native protein, e.g. its specific antigenic property. "Variants” of proteins or peptides as defined in the context of the present invention may comprise conservative amino acid substitution(s) compared to their native, i.e. non-mutated physiological, sequence. Those amino acid sequences as well as their encoding nucleotide sequences in particular fall under the term variants as defined herein.
  • amino acids which originate from the same class, are exchanged for one another are called conservative substitutions.
  • these are amino acids having aliphatic side chains, positively or negatively charged side chains, aromatic groups in the side chains or amino acids, the side chains of which can enter into hydrogen bridges, e.g. side chains which have a hydroxyl function.
  • an amino acid having a polar side chain is replaced by another amino acid having a likewise polar side chain, or, for example, an amino acid characterized by a hydrophobic side chain is substituted by another amino acid having a likewise hydrophobic side chain (e.g.
  • Insertions and substitutions are possible, in particular, at those sequence positions which cause no modification to the three-dimensional structure or do not affect the binding region. Modifications to a three-dimensional structure by insertion(s) or deletion(s) can easily be determined e.g. using CD spectra (circular dichroism spectra) (Urry, 1985, Absorption, Circular Dichroism and ORD of Polypeptides, in: Modern Physical Methods in Biochemistry, Neuberger et a/, (ed.), Elsevier, Amsterdam).
  • variants of proteins or peptides as defined herein, which may be encoded by a nucleic acid molecule may also comprise those sequences, wherein nucleotides of the nucleic acid are exchanged according to the degeneration of the genetic code, without leading to an alteration of the respective amino acid sequence of the protein or peptide, i.e. the amino acid sequence or at least part thereof may not differ from the original sequence in one or more mutation(s) within the above meaning.
  • nucleic acid sequences or amino acid sequences as defined herein preferably the amino acid sequences encoded by a nucleic acid sequence of the polymeric carrier as defined herein or the amino acid sequences themselves
  • the sequences can be aligned in order to be subsequently compared to one another. Therefore, e.g. a position of a first sequence may be compared with the corresponding position of the second sequence. If a position in the first sequence is occupied by the same component (residue) as is the case at a position in the second sequence, the two sequences are identical at this position. If this is not the case, the sequences differ at this position.
  • the percentage to which two sequences are identical is then a function of the number of identical positions divided by the total number of positions including those positions which are only occupied in one sequence.
  • the percentage to which two sequences are identical can be determined using a mathematical algorithm.
  • a preferred, but not limiting, example of a mathematical algorithm which can be used is the algorithm of Karlin et al. (1993), PNAS USA, 90:5873-5877 or Altschul et a/. (1997), Nucleic Acids Res., 25:3389-3402.
  • a "variant" of a protein or peptide may have at least 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% amino acid identity over a stretch of 10, 20, 30, 50, 75 or 100 amino acids of such protein or peptide.
  • a "variant" of a nucleic acid sequence may have at least 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% nucleotide identity over a stretch of 10, 20, 30, 50, 75 or 100 nucleotide of such nucleic acid sequence
  • Derivative of a protein or peptide A derivative of a peptide or protein is typically understood to be a molecule that is derived from another molecule, such as said peptide or protein.
  • a “derivative" of a peptide or protein also encompasses fusions comprising a peptide or protein used in the present invention.
  • the fusion comprises a label, such as, for example, an epitope, e.g., a FLAG epitope or a V5 epitope.
  • the epitope is a FLAG epitope.
  • a tag is useful for, for example, purifying the fusion protein.
  • Fusion protein is typically an artificial peptide or protein. Fusion proteins are typically created through the joining of two or more open reading frames which originally coded for separate peptides or proteins wherein joining may optionally occur via a linker sequence. These joined open reading frames are typically translated in a single peptide, polypeptide or protein with functional properties derived from each of the original proteins or peptides.
  • Fusion protein does not relate to the terms "Fusion (F) protein” or F protein, which instead refer to a specific class of viral proteins (see above).
  • Humoral immunity refers typically to antibody production and the accessory processes that may accompany it.
  • a humoral immune response may be typically characterized, e.g., by Th2 activation and cytokine production, germinal center formation and isotype switching, affinity maturation and memory cell generation.
  • Humoral immunity also typically may refer to the effector functions of antibodies, which include pathogen and toxin neutralization, classical complement activation, and opsonin promotion of phagocytosis and pathogen elimination.
  • an immunogen is preferably a protein or peptide, e.g. the product of an in vivo translation of a provided antigenic function.
  • an immunogen may elicit at least or exclusively an adaptive immunogen/antigen-specific immune response.
  • an immunogen may in particular be a (F) protein of the virus family Paramyxoviridae and (e.g. artificial) variants thereof as well as immunogenic fragments of said Fusion (F) protein and respective variants.
  • an immunogen may also in particular be a Hemagglutinin (HA) protein of the virus family Orthomyxoviridae and (e.g. artificial) variants thereof as well as immunogenic fragments of said Hemagglutinin (HA) protein and respective variants.
  • HA Hemagglutinin
  • Immune response may typically either be a specific reaction of the adaptive immune system to a particular antigen (so called specific or adaptive immune response) or an unspecific reaction of the innate immune system (so called unspecific or innate immune response).
  • the invention relates to the core to specific reactions (adaptive immune responses) of the adaptive immune system. Particularly, it relates to adaptive immune responses to infections by viruses like e.g. RSV or influenza. However, this specific response can be supported by an additional unspecific reaction (innate immune response). Therefore, the invention also relates to a compound for simultaneous stimulation of the innate and the adaptive immune system to evoke an efficient adaptive immune response.
  • the immune system may protect organisms from infection. If a pathogen breaks through a physical barrier of an organism and enters this organism, the innate immune system provides an immediate, but non-specific response. If pathogens evade this innate response, vertebrates possess a second layer of protection, the adaptive immune system. Here, the immune system adapts its response during an infection to improve its recognition of the pathogen. This improved response is then retained after the pathogen has been eliminated, in the form of an immunological memory, and allows the adaptive immune system to mount faster and stronger attacks each time this pathogen is encountered. According to this, the immune system comprises the innate and the adaptive immune system. Each of these two parts contains so called humoral and cellular components.
  • Immunostimulatory RNA in the context of the invention may typically be a RNA that is able to induce an innate immune response itself. It usually does not have an open reading frame and thus does not provide a pepti de-antigen or immunogen but elicits an innate immune response e.g. by binding to a specific kind of Tolllike-receptor (TLR) or other suitable receptors. However, of course also mRNAs having an open reading frame and coding for a pepti de/protein (e.g. an antigenic function) may induce an innate immune response.
  • TLR Tolllike-receptor
  • the innate immune system also known as non-specific immune system, comprises the cells and mechanisms that defend the host from infection by other organisms in a non-specific manner. This means that the cells of the innate system recognize and respond to pathogens in a generic way, but unlike the adaptive immune system, it does not confer long-lasting or protective immunity to the host.
  • the innate immune system may be e.g. activated by ligands of pathogen-associated molecular patterns (PAMP) receptors, e.g.
  • PAMP pathogen-associated molecular patterns
  • TLRs Toll-like receptors
  • auxiliary substances such as lipopolysaccharides, TNF-alpha, CD40 ligand, or cytokines, monokines, lymphokines, interleukins or chemokines, IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-1 0, IL-12, IL- 13, IL-14, IL-1 5, IL-1 6, IL-1 7, IL-1 8, IL-19, IL-20, IL-21 , IL-22, IL-23, IL-24, IL-25, IL-26, IL- 27, IL-28, IL-29, IL-30, IL-31 , IL-32, IL-33, IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G- CSF, M-CSF, LT-beta, TNF-alpha, growth factors, and
  • a response of the innate immune system includes recruiting immune cells to sites of infection, through the production of chemical factors, including specialized chemical mediators, called cytokines; activation of the complement cascade; identification and removal of foreign substances present in organs, tissues, the blood and lymph, by specialized white blood cells; activation of the adaptive immune system through a process known as antigen presentation; and/or acting as a physical and chemical barrier to infectious agents.
  • a monocistronic RNA may typically be a RNA, preferably a mRNA, that encodes only one open reading frame.
  • An open reading frame in this context is a sequence of several nucleotide triplets (codons) that can be translated into a peptide or protein.
  • Nucleic acid The term nucleic acid means any DNA- or RNA-molecule and is used synonymous with polynucleotide. Wherever herein reference is made to a nucleic acid or nucleic acid sequence encoding a particular protein and/or peptide, said nucleic acid or nucleic acid sequence, respectively, preferably also comprises regulatory sequences allowing in a suitable host, e.g. a human being, its expression, i.e. transcription and/or translation of the nucleic acid sequence encoding the particular protein or peptide.
  • a peptide is a polymer of amino acid monomers. Usually the monomers are linked by peptide bonds.
  • the term "peptide" does not limit the length of the polymer chain of amino acids. In some embodiments of the present invention a peptide may for example contain less than 50 monomer units. Longer peptides are also called polypeptides, typically having 50 to 600 monomeric units, more specifically 50 to 300 monomeric units.
  • Pharmaceutically effective amount A pharmaceutically effective amount in the context of the invention is typically understood to be an amount that is sufficient to induce an immune response.
  • a protein typically consists of one or more peptides and/or polypeptides folded into 3-dimensional form, facilitating a biological function.
  • a poly-(C )-sequence is typically a long sequence of cytosine nucleotides, typically about 10 to about 200 cytosine nucleotides, preferably about 10 to about 100 cytosine nucleotides, more preferably about 10 to about 70 cytosine nucleotides or even more preferably about 20 to about 50 or even about 20 to about 30 cytosine nucleotides.
  • a poly(C) sequence may preferably be located 3' of the coding region comprised by a nucleic acid.
  • a poly-A-tail also called "3'-poly(A) tail” is typically a long sequence of adenosine nucleotides of up to about 400 adenosine nucleotides, e.g. from about 25 to about 400, preferably from about 50 to about 400, more preferably from about 50 to about 300, even more preferably from about 50 to about 250, most preferably from about 60 to about 250 adenosine nucleotides, added to the 3' end of a RNA.
  • Polyadenylation is typically the addition of a Poly-A-Tail to a RNA, particularly to an mRNA. It is induced by a so called polyadenylation signal.
  • This signal may be typically located at the 3'-end of a RNA to be polyadenylated and may typically comprise a hexamer consisting of adenine and uracil, preferably the hexamer AAUAAA. Other hexamer sequences are conceivable.
  • Stabilized nucleic acid A stabilized nucleic acid, typically, exhibits a modification increasing resistance to in vivo degradation (e.g. degradation by an exo- or endo-nuclease) and/or ex vivo degradation (e.g. by the manufacturing process prior to vaccine administration, e.g. in the course of the preparation of the vaccine solution to be administered).
  • Stabilization of RNA can, e.g., be achieved by providing a 5'Cap-Structure, a Poly-A-Tail, or any other UTR-modification. It can also be achieved by backbone- modification or modification of the G/C-content of the nucleic acid.
  • Vaccine A vaccine is typically understood to be a prophylactic or therapeutic material providing at least one antigenic function, particularly an immunogen. The antigen or immunogen stimulates the body's adaptive immune system to provide an adaptive immune response.
  • An agent e.g. a carrier, that may typically be used within a vaccine for facilitating administering of the immunogenic composition and or the antigenic function to an individual.
  • the invention provides a combination vaccine providing at least a first and a second antigenic function; the combination vaccine comprising at least one RNA (preferably mRNA) encoding at least one or more proteins or fragments, variants or derivatives of proteins awarding the antigenic functions; wherein the first antigenic function being a Fusion (F) protein or a fragment, variant or derivative of a Fusion (F) protein derived from the virus family Paramyxoviridae and the second antigenic function being an Hemagglutinin (HA) protein or a fragment, variant or derivative of an Hemagglutinin (HA) protein derived from the virus family Orthomyxoviridae.
  • RNA preferably mRNA
  • each RNA encoding an antigenic function is an antigen- providing RNA according to the above given definition.
  • the immuno-active component (that means the component that causes an interaction with the immune system of the treated individual to provoke preferably an adaptive immune response) is at least one antigen- providing RNA.
  • the combination vaccine can contain either one antigen- providing RNA that encodes both or all antigenic functions or two or more distinct antigen- providing RNAs encoding both or all antigenic functions.
  • the RNA in the combination vaccine may for example be an mRNA.
  • the RNA may be a virus derived RNA vector such as a retrovirus or an alphavirus derived RNA replicon vector.
  • a retrovirus is an RNA virus that is duplicated in a host cell using the reverse transcriptase enzyme to produce DNA from its RNA genome. The DNA is then incorporated into the host's genome by an integrase enzyme. The virus thereafter replicates as part of the host cell's DNA and then undergoes the usual transcription and translational processes to express the genes carried by the virus.
  • Alphaviruses are single stranded RNA viruses in which heterologous genes of interest may substitute for the alphavirus' structural genes.
  • the replicon RNA is packaged into replicon particles (RP) which may be used for example for vaccination (see for example Vander Veen et a/., 2012. Alphavirus replicon vaccines. Animal Health Research Reviews, p. 1 -9).
  • RP replicon particles
  • the genomic viral RNA After entry into the host cell, the genomic viral RNA initially serves as an mRNA for translation of the viral nonstructural proteins (nsPs) required for initiation of viral RNA amplification.
  • nsPs viral nonstructural proteins
  • RNA replication occurs via synthesis of a full-length minusstrand intermediate that is used as the template for synthesis of additional genome-length RNAs and for transcription of a plus- strand subgenomic RNA from an internal promoter. Such RNA may then be considered as self replicating RNA, since the non-structural proteins responsible for replication (and transcription of the heterologous genes) are stil l present in such replicon.
  • the Fusion (F) protein and the Hemagglutinin (HA) protein as encoded by said RNA e.g. mRNA or viral RNA
  • RNA e.g. mRNA or viral RNA
  • the encoded proteins or fragments, variants or derivatives of the proteins may be antigens, particularly immunogens. It is also possible that the coding sequence of the Fusion (F) protein and/or the Hemagglutinin (HA) protein is distributed over two or more RNAs and/or over two or more open reading frames. The two or more RNAs and/or two or more open reading frames will in such scenario encode several distinct fragments of the Fusion (F) protein and/or the Hemagglutinin (HA) protein.
  • the inventive combination vaccine comprising at least one RNA providing these antigenic functions (HA protein and F protein, or fragments, variants or derivatives thereof) does show an unexpectedly remarkable synergistic effect.
  • a combination vaccine comprising RNAs encoding a Fusion (F) protein of the virus family Paramyxoviridae, particularly RSV and a Hemagglutinin (HA) protein of the virus family Orthomyxoviridae, particularly Influenza, or fragments, variants or derivatives thereof, provides an improved Fusion (F) protein-specific immune response, particularly a superior specific T cell response compared to vaccination with mRNA coding solely for the Fusion (F) protein.
  • the combination vaccine according to the invention is thus preferably suitable to elicit an antigen-specific immune response in a patient.
  • the mRNA encoded Fusion (F) protein and Hemagglutinin (HA) protein serve as antigens.
  • the RNA encoding the Fusion (F) protein or a fragment, variant or derivative thereof of the virus family Paramyxoviridae and the RNA encoding the Hemagglutinin (HA) protein or a fragment, variant or derivative thereof of the virus family Orthomyxoviridae are comprised in the same composition of the combination vaccine.
  • Quasi-simultaneous administration may, alternatively, be also achieved by subsequent administration (within e.g. up to 1 0 minutes, more preferably within two minutes) of a combination vaccine which is composed of e.g.
  • the first composition contains RNA encoding the Fusion (F) protein or a fragment, variant or derivative thereof of the virus family Paramyxoviridae
  • the second composition contains the RNA encoding the Hemagglutinin (HA) protein or a fragment, variant or derivative thereof of the virus family Orthomyxoviridae.
  • both compositions it is preferred to administer both compositions at the same site of the body or at least close to each other such that the same area of the patient's lymphatic system is addressed by both administrations, thereby triggering an immune response which as coherent as an immune response triggered by the administration of a combination vaccine composed one single composition containing mRNA molecules encoding both antigenic functions.
  • a "staggered" combination vaccine may, alternatively, be provided by subsequent administration by separate compositions, each composition comprising distinct immunogens and/or antigenic functions.
  • a immune response is to be triggered which is comparable to the coherent immune response achieved by the administration of one single composition, i.e. the synergistic effect on e.g. the immune response against the F protein.
  • this approach according to the invention shows the potential of an RNA based vaccine allowing simultaneous vaccination against viruses belonging to the virus families Paramyxoviridae and Orthomyxoviridae, respectively, by combination of RNA vaccines encoding relevant viral antigens.
  • the combination of RNAs encoding the Fusion (F) protein or a fragment, variant or derivative thereof of e.g. RSV strains and the Hemagglutinin (HA) protein or a fragment, variant or derivative thereof of e.g. Influenza viruses was shown to specifically enhance the adaptive immune response against the e.g. RSV F protein in an unexpected way.
  • the combination vaccine according to the invention provides not only a mixture of RNAs encoding different antigens (of two distinct viruses) but also an unexpected synergistic effect for the F protein specific T cell immune response.
  • Any functional fragment, variant or derivative of the Fusion (F) protein or the Hemagglutinin (HA) protein, which may be encoded by the RNAs of the inventive combination vaccine shall advantageously trigger the same synergistic immune response as the corresponding full-length proteins, in particular the same specific T cell immune response and preferably also the same B-cell response, as observed for the full-length protein-based combination vaccine, against the F protein of e.g. RSV.
  • the "same” in this regard means of "the same order of magnitude”.
  • any functional fragment, variant or derivative of the full- length F or HA proteins contains the decisive epitopes of the full-length HA or F protein sequences such that the immune response is not decreased due to less antigenic potential of the fragments, variant or derivative.
  • the antigenic functions are provided by the combination vaccine in the form of monocistronic RNAs, whereby a first monocistronic RNA encodes said Fusion (F) protein or said fragment, variant or derivative thereof and a second monocistronic RNA encodes said Hemagglutinin (HA) protein or said fragment, variant or derivative thereof.
  • the antigenic functions are provided by the combination vaccine in the form of a bicistronic or a multicistronic RNA.
  • the bi- or multicistronic RNA may contain at least one open reading frame, which encodes said Fusion (F) protein or said fragment, variant or derivative thereof and wherein at least one other open reading frame encodes said Hemagglutinin (HA) protein or said fragment, variant or derivative thereof.
  • both antigenic functions are provided by one single RNA molecule.
  • such a bi- or multicistronic RNA may encode, e.g., two or even more coding sequences of at least two antigenic functions, as defined above.
  • a bi- or multicistronic RNA may e.g.
  • RNA may, e.g., contain distinct antigenic functions of the Hemagglutinin (HA) protein (derived e.g. from the same or from different Influenza strains).
  • F Fusion
  • HA Hemagglutinin
  • the combination vaccine comprises a first bi- or multicistronic RNA encoding for an ensemble of Fusion (F) proteins or fragments, variants or derivatives thereof derived from different Paramyxoviridae and a second monocistronic RNA encoding for a Hemagglutinin (HA) protein derived from a virus belonging to the Orthomyxoviridae, or the other way around.
  • F Fusion
  • HA Hemagglutinin
  • the coding sequences of such bi- or multicistronic RNAs may be separated by at least one internal ribosomal entry site (IRES) sequence.
  • IRES sequence can function as a sole ribosome binding site, but it can also serve to provide a bi- or even multicistronic RNA as defined herein which codes for several antigens, which are to be translated by the ribosomes independently of one another.
  • IRES sequences which can be used according to the invention are those from picornaviruses (e.g.
  • FMDV pestiviruses
  • CFFV pestiviruses
  • PV polioviruses
  • ECMV encephalomyocarditis viruses
  • FMDV foot and mouth disease viruses
  • HCV hepatitis C viruses
  • CSFV classical swine fever viruses
  • MLV mouse leukemia virus
  • SIV simian immunodeficiency viruses
  • CrPV cricket paralysis viruses
  • the antigenic functions are provided by the combination vaccine in the form of a monocistronic RNA encoding the Fusion (F) protein or a fragment, variant or derivative thereof and encoding the Hemagglutinin (HA) protein or a fragment, variant or derivative thereof as a fusion protein.
  • a fusion protein e.g. the full-length sequences of the Fusion (F) protein and the full- length sequence of the Hemagglutinin (HA) protein are linked with or without a linker sequence.
  • such a fusion protein may contain a full-length protein sequence of the Fusion (F) protein and only parts of the Hemagglutinin (HA) protein (or vice versa) or may contain parts of either both of these proteins.
  • RNAs encoding fusion proteins which are composed of one or more antigenic peptide sequences, encoding epitopes of the Fusion (F) and/or the Hemagglutinin (HA) protein that can individually act as immunogens.
  • each of these proteins are preferably arranged in a non- native way, which means that the epitope sequences are isolated from the native sequences and are linked by non-native linker sequences (e.g linker sequences having more than 50% glycine and proline residues).
  • linker sequences e.g linker sequences having more than 50% glycine and proline residues.
  • inventive monocistronic RNAs encoding such fusion proteins may be provided with or without linker sequences.
  • linker sequences typically comprise 5 to 25 amino acids, preferably selected from proline and glycine.
  • the linker sequence is immunologically neutral, e.g. non-immunogenic and non-immunostimulatory.
  • the at least one Fusion (F) protein is derived from viruses selected from: Avulavirus, Ferlavirus, Henipavirus, Morbillivirus, Respirovirus, Rubulavirus, TPMV-like viruses, Pneumovirus, Metapneumovirus, Atlantic salmon paramyxovirus, Beilong virus, J virus, Mossman virus, Nariva virus, Salem virus, or Pacific salmon paramyxovirus.
  • Avulavirus can be e.g. Newcastle disease virus; Ferlavirus can be e.g. Fer-de-Lance virus; Henipavirus can be e.g. Hendravirus, Nipahvirus; Morbillivirus can be e.g.
  • Respirovirus can be e.g. Sendai virus, Human Parainfluenza viruses 1 and 3, viruses of the common cold; Rubulavirus can be e.g. Mumps virus, Human Parainfluenza viruses 2 and 4, Simian Parainfluenza virus 5, Menangle virus, Tioman virus, Tuhokovirus 1, 2 and 3; TPMV-like viruses can be e.g. Tupaia paramyxovirus; Pneumovirus can be e.g.
  • the Fusion (F) protein is derived from human respiratory syncytial virus (RSV), preferably selected from RSV Long (preferably according to SEQ ID No. 1 ) or RSV A2 (preferably according to SEQ ID No. 2 or mutants thereof such as P102A, 1379V or M447V), more preferably the Fusion (F) protein is a protein encoded at least partially by one of the nucleic acid sequences according to SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 7, SEQ ID No. 8, SEQ ID No.
  • the combination vaccine of the invention can contain an ensemble of more than one antigenic function derived from distinct Fusion (F) proteins, which may either be derived from distinct strains of e.g. the above viruses or derived from (e.g. the above) different viruses or may be a combination of both. They may be provided distinct RNA molecules (more than one type) or by a single RNA molecule (one type).
  • F Fusion
  • the distinct antigenic functions may be provided by a monocistronic type of RNA encoding a fusion protein presenting these distinct antigenic functions or by a bi- or multicistronic RNA coding for distinct antigenic functions.
  • a monocistronic type of RNA encoding a fusion protein presenting these distinct antigenic functions
  • a bi- or multicistronic RNA coding for distinct antigenic functions may be combined and do not exclude each other.
  • the at least one Hemagglutinin (HA) protein is derived from an Influenza virus, preferably selected from: Influenza A (e.g. H1 N1, H1 N2, H2N2, H3N1 , H3N2, H3N8, H5N1, H5N2, H5N3, H5N8, H5N9, H7N1 , H7N2, H7N3, H7N4, H7N7, H9N1 , H9N2, H10N7), Influenza B, Influenza C, Isavirus (e.g. Infectious salmon anemia virus), Thogotovirus (e.g.
  • the HA protein is a protein according to SEQ ID No. 3, more preferably the Hemagglutinin (HA) protein is a protein encoded at least partially by the nucleic acid sequence according to SEQ ID No. 6, SEQ ID No: 12, SEQ ID No: 18, or SEQ ID No. 21 . More preferably, the HA protein as encoded by any of the above SEQ ID Nos. may be combined, e.g. for providing one single composition comprising at least two nucleic acids, e.g. SEQ ID No. 18 or SEQ ID No. 21 , with a F protein encoded by any of the following SEQ ID Nos. 13, 14, 15, 16, 1 7.
  • SEQ ID No. 18 or SEQ ID No. 21 may be combined for the combination vaccine, e.g. in the form of one single composition or as a staggered combination vaccine, with SEQ ID No. 13, alternatively, with SEQ ID No. 14. or alternatively with SEQ ID No. 15, or alternatively with SEQ ID No: 16, or alternatively SEQ ID No. 17 or alternatively SEQ ID No 19 or alternatively SEQ ID No 20.
  • the combination vaccine of the invention can contain an ensemble of more than one antigenic function derived from distinct Hemagglutinin (HA) proteins, which may either be derived from distinct strains of e.g. the above viruses or derived from (e.g. the above) different viruses or may be a combination of both. They may be provided by more distinct RNA molecules (more than one type) or by a single RNA molecule (one type). If provided by one single RNA type, the distinct antigenic functions may be provided by a monocistronic type of RNA encoding a fusion protein presenting these distinct antigenic functions or a bi- or multicistronic RNA coding for distinct antigenic functions. Of course, the above embodiments may be combined and do not exclude each other.
  • HA Hemagglutinin
  • the at least one RNA of the inventive combination vaccine may be stabilized in order to prevent instability and (fast) degradation of the RNA (or any further nucleic acid molecule) by various approaches.
  • This instability of RNA is typically due to RNA-degrading enzymes, "RNases" (ribonucleases), wherein contamination with such ribonucleases may sometimes completely degrade RNA in solution.
  • RNases ribonucleases
  • RNases ribonucleases
  • the natural degradation of RNA in the cytoplasm of cells is very finely regulated and RNase contaminations may be generally removed by special treatment prior to use of said compositions, in particular with diethyl pyrocarbonate (DEPC).
  • DEPC diethyl pyrocarbonate
  • the terminal structure is typically of critical importance particularly for an mRNA.
  • a so-called cap structure which is a modified guanosine nucleotide also called 5'Cap structure, and at the 3' end is typically a sequence of up to 200 adenosine nucleotides (the so-called poly-A tail).
  • the at least one RNA comprises at least one of the following structural elements: a histone-stem-loop structure, preferably a histone-stem-loop in its 3' untranslated region, a 5'Cap structure, a poly(C) sequence, a poly-A tail and/or a polyadenylation signal, preferably as defined herein.
  • the at least one RNA preferably comprises at least two of the following structural elements: a 5' and/or 3'-stabilizing sequence; a histone-stem-loop structure, preferably a histone-stem-loop in its 3' untranslated region; a 5'-Cap structure; a poly(C) sequence; a poly-A tail; or a polyadenylation signal, e.g. given a 5'-Cap structure and a histone-stem-loop and, potentially a poly-A-tail.
  • a 5' and/or 3'-stabilizing sequence e.g. given a 5'-Cap structure and a histone-stem-loop and, potentially a poly-A-tail.
  • Stabilizing sequences in the 5' and/or 3' untranslated regions have the effect of increasing the half-life of the nucleic acid in the cytosol.
  • These stabilizing sequences can have 100% sequence identity to naturally occurring sequences which occur in viruses, bacteria and eukaryotes, but can also be partly or completely synthetic.
  • the untranslated sequences (UTR) of the (alpha-)globin gene e.g. from Homo sapiens or Xenopus laevis may be mentioned as an example of stabilizing sequences which can be used in the present invention for a stabilized nucleic acid.
  • stabilizing sequence has the general formula (QU)CCAN x CCC(U/A)Py x UC(aU)CC) which is contained in the 3' UTR of the very stable RNA which codes for (alpha-)globin, type(l)-collagen, 15-lipoxygenase or for tyrosine hydroxylase (cf. Holcik et a/., Proc. Natl. Acad. Sci. USA 1997, 94: 241 0 to 2414).
  • Such stabilizing sequences can of course be used individually or in combination with one another and also in combination with other stabilizing sequences known to a person skilled in the art.
  • a histone stem-loop sequence suitable to be used within the present invention, is preferably selected from at least one of the following formulae (I) or (II): formula (I) (stem-loop sequence without stem bordering elements):
  • steml loop stem2 formula (II) (stem-loop sequence with stem bordering elements): [N 0 .4(U/T)No, 4 ] [N3- 5 CNo. 2 ]
  • steml steml loop stem2 stem2 bordering element bordering element wherein: steml or stem2 bordering elements N, .6 is a consecutive sequence of 1 to 6, preferably of 2 to 6, more preferably of 2 to 5, even more preferably of 3 to 5, most preferably of 4 to 5 or 5 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C, or a nucleotide analogue thereof;
  • steml is reverse complementary or partially reverse complementary with element stem2, and is a consecutive sequence between of 5 to 7 nucleotides; wherein N 0 . 2 is a consecutive sequence of 0 to 2, preferably of 0 to 1 , more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof; wherein N 3 .
  • s is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof, and wherein G is guanosine or an analogue thereof, and may be optionally replaced by a cytidine or an analogue thereof, provided that its complementary nucleotide cytidine in stem2 is replaced by guanosine; loop sequence [NWU/TJNQ- is located between elements steml and stem2, and is a consecutive sequence of 3 to 5 nucleotides, more preferably of 4 nucleotides; wherein each N 0 .
  • each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof;
  • U/T represents uridine, or optionally thymidine
  • stem2 [N 3 . 5 CN 0 . 2 ] is reverse complementary or partially reverse complementary with element steml , and is a consecutive sequence between of 5 to 7 nucleotides; wherein N 3 .
  • 5 is a consecutive sequence of 3 to 5, preferably of 4 to 5, more preferably of 4 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G and C or a nucleotide analogue thereof;
  • No_ 2 is a consecutive sequence of 0 to 2, preferably of 0 to 1 , more preferably of 1 N, wherein each N is independently from another selected from a nucleotide selected from A, U, T, G or C or a nucleotide analogue thereof; and wherein C is cytidine or an analogue thereof, and may be optionally replaced by a guanosine or an analogue thereof provided that its complementary nucleoside guanosine in steml is replaced by cytidine; wherein
  • steml and stem2 are capable of base pairing with each other forming a reverse complementary sequence, wherein base pairing may occur between steml and stem2, e.g. by Watson-Crick base pairing of nucleotides A and U/T or G and C or by non-Watson-Crick base pairing e.g. wobble base pairing, reverse Watson-Crick base pairing, Hoogsteen base pairing, reverse Hoogsteen base pairing or are capable of base pairing with each other forming a partially reverse complementary sequence, wherein an incomplete base pairing may occur between steml and stem2, on the basis that one ore more bases in one stem do not have a complementary base in the reverse complementary sequence of the other stem.
  • a further optionally comprised stabilizing element is a poly(A) sequence, also called poly-A- tail at the 3'-terminus of the at least one RNA.
  • a poly(A) sequence comprises a sequence of about 25 to about 400 adenosine nucleotides, preferably a sequence of about 50 to about 400 adenosine nucleotides, more preferably a sequence of about 50 to about 300 adenosine nucleotides, even more preferably a sequence of about 50 to about 250 adenosine nucleotides, most preferably a sequence of about 60 to about 250 adenosine nucleotides.
  • the term "about” refers to a deviation of ⁇ 10% of the value(s) it is attached to.
  • the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine as defined herein can be modified by a sequence of at least 10 cytosines, preferably at least 20 cytosines, more preferably at least 30 cytosines (so-called "poly(C) sequence").
  • the nucleic acid molecule may contain or code for a poly(C) sequence of typically about 10 to 200 cytosine nucleotides, preferably about 10 to 100 cytosine nucleotides, more preferably about 10 to 70 cytosine nucleotides or even more preferably about 20 to 50 or even 20 to 30 cytosine nucleotides.
  • This poly(C) sequence is preferably located 3' of the coding region comprised in the nucleic acid particularly in the at least one RNA according to the first aspect of the present invention.
  • the at least one RNA encoding at least one Fusion (F) protein or a fragment, variant or derivative thereof of the virus fami ly Paramyxoviridae and at least one Hemagglutinin (HA) protein or a fragment, variant or derivative thereof of the virus family Orthomyxoviridae has the following structure in 5' to 3'- direction: a) a coding region, preferably encoding a peptide or protein as defined above;
  • the at least one RNA encoding at least one Fusion (F) protein or a fragment, variant or derivative thereof of the virus fami ly Paramyxoviridae and at least one Hemagglutinin (HA) protein or a fragment, variant or derivative thereof of the virus family Orthomyxoviridae (or any other coding nucleic acid comprised in the inventive combination vaccine) has the following structure in 5' to 3'-direction: a) a coding region, preferably encoding a peptide or protein as defined above;
  • the coding region might be or might comprise at least partially the coding region of one of the sequences according to SEQ ID No. 4 to SEQ ID No. 21 .
  • the RNA might be or might comprise at least partially one of the sequences according to SEQ ID No. 4 to SEQ ID No. 21 .
  • the RNA e.g. mRNA
  • the RNA might comprise a combination of at least two of these sequences or a combination of fragments or variants thereof.
  • at least one sequence is preferably selected from SEQ ID No. 1 9; or SEQ ID No. 20, and a further sequence is preferably selected from SEQ ID No. 21 .
  • Other combinations can of course be imagined as well.
  • the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine may be provided as a stabilized nucleic acid, e.g. in the form of a modified nucleic acid.
  • the at least one RNA or any further nucleic acid comprised in the inventive combination vaccine is stabilized, preferably by backbone modifications, sugar modifications and/or base modifications, more preferred stabilized by modification of the G/C-content.
  • RNA modifications may be introduced into the at least one RNA without impairing the RNA's function to be translated into the antigenic function derived from the Fusion (F) protein or the Hemagglutinin (HA) protein or any further encoded protein or peptide.
  • F Fusion
  • HA Hemagglutinin
  • a backbone modification in the context of the present invention is preferably a modification in which phosphates of the backbone of the nucleotides contained in the at least one RNA of the inventive combination vaccine (or any further nucleic acid as defined herein) are chemically modified, e.g. anionic internucleoside linkage, N3'- P5' modifications, replacement of non-bridging oxygen atoms by boranes, neutral internucleoside linkage, amide linkage of the nucleosides, methylene(methylimino) linkages, formacetal and thioformacetal linkages, introduction of sulfonyl groups, or the like.
  • anionic internucleoside linkage e.g. anionic internucleoside linkage, N3'- P5' modifications, replacement of non-bridging oxygen atoms by boranes, neutral internucleoside linkage, amide linkage of the nucleosides, methylene(methylimino) linkages, formacetal and
  • a sugar modification in the context of the present invention is preferably a chemical modification of the sugar of the nucleotides of the at least one RNA of the inventive combination vaccine (or any further nucleic acid as defined herein), e.g. methylation of the ribose residue or the like.
  • the at least one RNA of the inventive combination vaccine or any further coding nucleic acid comprised in the inventive combination vaccine may be modified and thus stabi lized by modifying the G (guanosine)/C (cytosine) content of the mRNA, preferably of the coding region thereof.
  • the G/C content of the at least one RNA of the inventive combination vaccine or any further coding nucleic acid comprised in the inventive combination vaccine is particularly increased compared to the G/C content of the coding region of its particular wild type coding sequence, i.e. the unmodified RNA.
  • the encoded amino acid sequence of the RNA or coding nucleic acid is preferably not modified compared to the coded amino acid sequence of the particular wild type RNA or coding nucleic acid.
  • the modification of the G/C-content of the at least one RNA of the inventive combination vaccine or any further coding nucleic acid comprised in the inventive combination vaccine is based on the fact that RNA sequences having an increased G (guanosine)/C (cytosine) content are more stable than RNA sequences having an increased A (adenosine)/U (uracil) content.
  • the codons of a coding sequence or a whole RNA might therefore be varied compared to the wild type coding sequence or mRNA, such that they include an increased amount of G/C nucleotides while the translated amino acid sequence is retained.
  • the G/C content of the coding region of the at least one RNA of the inventive combination vaccine or any further coding nucleic acid comprised in the inventive combination vaccine according to the invention is increased by at least 7%, more preferably by at least 15%, particularly preferably by at least 20%, compared to the G/C content of the coded region of the wild type RNA.
  • At least 5%, 10%, 20%, 30%, 40%, 50%, 60%, more preferably at least 70 %, even more preferably at least 80% and most preferably at least 90%, 95% or even 100% of the substitutable codons in the region coding for a protein or peptide as defined herein or its fragment or variant thereof or the whole sequence of the wild type RNA sequence or coding sequence are substituted, thereby increasing the G/C content of said sequence.
  • the at least one RNA or any further coding nucleic acid comprised in the inventive combination vaccine is optimized for translation, preferably optimized for translation by replacing codons for less frequent tRNAs of a given amino acid by codons for more frequently occurring tRNAs of the respective amino acid. This is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells.
  • the corresponding modified RNA is translated to a significantly poorer degree than in the case where codons coding for more frequent tRNAs are present.
  • the coding region of the at least one RNA of the inventive combination vaccine or any further coding nucleic acid comprised in the inventive combination vaccine is modified compared to the corresponding region of the wild type RNA or coding sequence such that at least one codon of the wild type sequence which codes for a tRNA which is relatively rare or less frequent in the cell is exchanged for a codon which codes for a tRNA which is more or most frequent in the cell and carries the same amino acid as the relatively rare or less frequent tRNA.
  • the sequences of the at least one RNA of the inventive combination vaccine or any further coding nucleic acid comprised in the inventive combination vaccine can be modified such that codons for which more frequently occurring tRNAs are available are inserted.
  • Substitutions, additions or eliminations of bases are preferably carried out using a DNA matrix for preparation of the nucleic acid molecule by techniques of the well known site directed mutagenesis or with an oligonucleotide ligation.
  • a corresponding DNA molecule may be transcribed in vitro.
  • This DNA matrix preferably comprises a suitable promoter, e.g. a T7 or SP6 promoter, for in vitro transcription, which is followed by the desired nucleotide sequence for the at least one RNA to be prepared and a termination signal for in vitro transcription.
  • the DNA molecule which forms the matrix of the at least one RNA of interest, may be prepared by fermentative proliferation and subsequent isolation as part of a piasmid which can be replicated in bacteria.
  • Plasmids which may be mentioned as suitable for the present invention are e.g. the plasmids pT7Ts (GenBank accession number U26404; Lai et al., Development 1995, 1 21 : 2349 to 2360), pGEM ® series, e.g. pGEM ® -1 (GenBank accession number X65300; from Promega) and pSP64 (GenBank accession number X65327); cf. also Mezei and Storts, Purification of PCR Products, in: Griffin and Griffin (ed.), PCR Technology: Current Innovation, CRC Press, Boca Raton, FL, 2001 .
  • nucleic acid molecules used e.g. the at least one RNA of the inventive combination vaccine or any further nucleic acid molecule as defined herein, may be prepared using any method known in the art, including synthetic methods such as e.g. solid phase synthesis, as well as in vitro methods, such as in vitro transcription reactions.
  • the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine may be administered naked without being associated with any further vehicle, transfection or complexation agent for increasing the transfection efficiency and/or the immunostimulatory properties of the at least one RNA or of further comprised nucleic acid.
  • the at least one RNA of the inventive combination vaccine or any other nucleic acid comprised in the inventive combination vaccine according to the invention may be formulated together with a cationic or polycationic compound and/or with a polymeric carrier.
  • the at least one RNA or any other nucleic acid comprised in the inventive combination vaccine is associated with or complexed with a cationic or polycationic compound or a polymeric carrier, optionally in a weight ratio selected from a range of about 6:1 (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/w) to about 0.5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 : 1 (w/w) or of about 3:1 (w/w) to about 1 : 1 (w/w), and most preferably a ratio of about 3: 1 (w/w) to about 2:1 (w/w) of RNA to cationic or polycationic
  • the at least one RNA of the inventive combination vaccine or any other nucleic acid comprised in the inventive combination vaccine can also be associated with a vehicle, transfection or complexation agent for increasing the transfection efficiency and/or the immunostimulatory properties of the at least one RNA or of optionally comprised further included nucleic acids.
  • Cationic or polycationic compounds being particularly preferred agents in this context include protamine, nucleoline, spermine or spermidine, or other cationic peptides or proteins, such as poly-L-lysine (PLL), poly-arginine, basic polypeptides, cell penetrating peptides (CPPs), including HIV-binding peptides, HIV-1 Tat (HIV), Tat-derived peptides, Penetratin, VP22 derived or analog peptides, HSV VP22 (Herpes simplex), MAP, KALA or protein transduction domains (PTDs), PpT620, prolin-rich peptides, arginine-rich peptides, lysine-rich peptides, MPG-peptide(s), Pep-1 , L-oligomers, Calcitonin peptide(s), Antennapedia-derived peptides (particularly from Drosophila antennapedia), pAntp, plsl,
  • protamine is particularly preferred.
  • preferred cationic or polycationic proteins or peptides may be selected from the following proteins or peptides having the following total formula (III):
  • Particularly preferred cationic peptides in this context are e.g. Arg 7 , Arge, Arg 9 , H 3 R 9 , R 9 H 3 , H 3 R 9 H 3 , YSSR 9 SSY, (RKH) repeat, Y(RKH) 2 R, etc.
  • cationic or polycationic compounds which can be used as transfection or complexation agent may include cationic polysaccharides, for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEL), cationic lipids, e.g.
  • cationic polysaccharides for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEL), cationic lipids, e.g.
  • PEL polyethyleneimine
  • DOTMA [1 -(2,3- sioleyloxy)propyl)]-N,N,N-trimethylammonium chloride, DMRIE, di-C14-amidine, DOTIM, SAINT, DC-Choi, BGTC, CTAP, DOPC, DODAP, DOPE: Dioleyl phosphatidylethanol- amine, DOSPA, DODAB, DOIC, DMEPC, DOGS: Dioctadecylamidoglicylspermin, DIMRI: Dimyristo-oxypropyl dimethyl hydroxyethyl ammonium bromide, DOTAP: dioleoyloxy-3- (trimethylammonio)propane, DC-6-14: O,O-ditetradecanoyl-N-(a- trimethylammonioacetyl)diethanolamine chloride, CLIP1 : rac-[(2,3-dioctadecyloxypropyl)(2- hydroxyethyl)]-d
  • modified polyaminoacids such as ⁇ -aminoacid-polymers or reversed polyamides, etc.
  • modified polyethylenes such as PVP (poly(N-ethyl-4-vinylpyridinium bromide)), etc.
  • modified acrylates such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)), etc.
  • modified amidoamines such as pAMAM (poly(amidoamine)), etc.
  • modified polybetaaminoester (PBAE) such as diamine end modified 1 ,4 butanediol diacrylate-co-5-amino-1 -pentanol polymers, etc.
  • dendrimers such as polypropylamine dendrimers or pAMAM based dendrimers, etc.
  • polyimine(s) such as PEI: poly(ethyleneimine), poly(propyleneimine), etc.
  • polyallylamine sugar backbone based
  • a polymeric carrier according to the invention might be a polymeric carrier formed by disulfide-crosslinked cationic components.
  • the disulfide-crosslinked cationic components may be the same or different from each other.
  • the polymeric carrier can also contain further components. It is also particularly preferred that the polymeric carrier of the present invention comprises mixtures of cationic peptides, proteins or polymers and optionally further components as defined herein, which are crosslinked by disulfide bonds as described herein.
  • the cationic components which form basis for the polymeric carrier by disulfide-crosslinkage, are typically selected from any suitable cationic or polycationic peptide, protein or polymer suitable for this purpose, particular any cationic or polycationic peptide, protein or polymer capable to complex a nucleic acid as defined according to the present invention, and thereby preferably condensing the nucleic acid.
  • the cationic or polycationic peptide, protein or polymer is preferably a linear molecule, however, branched cationic or polycationic peptides, proteins or polymers may also be used.
  • Every disulfide-crosslinking cationic or polycationic protein, peptide or polymer of the polymeric carrier which may be used to complex the at least one RNA of the combination vaccine or any further nucleic acid comprised in the combination vaccine contains at least one -SH moiety, most preferably at least one cysteine residue or any further chemical group exhibiting an -SH moiety, capable to form a disulfide linkage upon condensation with at least one further cationic or polycationic protein, peptide or polymer as cationic component of the polymeric carrier as mentioned herein.
  • the polymeric carrier which may be used to complex the at least one RNA of the combination vaccine or any further nucleic acid comprised in the combination vaccine may be formed by disulfide-crosslinked cationic (or polycationic) components.
  • At least one cationic (or polycationic) component of the polymeric carrier which may be used to complex the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine may be selected from cationic or polycationic peptides or proteins.
  • Such cationic or polycationic peptides or proteins preferably exhibit a length of about 3 to 100 amino acids, preferably a length of about 3 to 50 amino acids, more preferably a length of about 3 to 25 amino acids, e.g. a length of about 3 to 10, 5 to 15, 10 to 20 or 15 to 25 amino acids.
  • such cationic or polycationic peptides or proteins may exhibit a molecular weight of about 0.01 kDa to about 100 kDa, including a molecular weight of about 0.5 kDa to about 100 kDa, preferably of about 10 kDa to about 50 kDa, even more preferably of about 10 kDa to about 30 kDa.
  • the cationic component of the polymeric carrier which may be used to complex the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine comprises a cationic or polycationic peptide or protein
  • the cationic properties of the cationic or polycationic peptide or protein or of the entire polymeric carrier if the polymeric carrier is entirely composed of cationic or polycationic peptides or proteins, may be determined upon its content of cationic amino acids.
  • the content of cationic amino acids in the cationic or polycationic peptide or protein and/or the polymeric carrier is at least 10%, 20%, or 30%, preferably at least 40%, more preferably at least 50%, 60% or 70%, but also preferably at least 80%, 90%, or even 95%, 96%, 97%, 98%, 99% or 100%, most preferably at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, or may be in the range of about 10% to 90%, more preferably in the range of about 15% to 75%, even more preferably in the range of about 20% to 50%, e.g.
  • such cationic or polycationic peptides or proteins of the polymeric carrier which comprise or are additionally modified to comprise at least one -SH moiety, are selected from, without being restricted thereto, cationic peptides or proteins such as protamine, nucleoline, spermine or spermidine, oligo- or poly-L-lysine (PLL), basic polypeptides, oligo or poly-arginine, cell penetrating peptides (CPPs), chimeric CPPs, such as Transportan, or MPG peptides, HIV-binding peptides, Tat, HIV-1 Tat (HIV), Tat-derived peptides, members of the penetratin family, e.g.
  • cationic peptides or proteins such as protamine, nucleoline, spermine or spermidine, oligo- or poly-L-lysine (PLL), basic polypeptides, oligo or poly-arginine, cell penetrating peptides (
  • Penetratin Antennapedia-derived peptides (particularly from Drosophila antennapedia), pAntp, plsl, etc., antimicrobial-derived CPPs e.g. Buforin-2, Bac71 5-24, SynB, SynB(l ), pVEC, hCT-derived peptides, SAP, MAP, PpTG20, Loligomere, FGF, Lactoferrin, histones, VP22 derived or analog peptides, Pestivirus Ems, HSV, VP22 (Herpes simplex), MAP, KALA or protein transduction domains (PTDs, PpT620, prol in-rich peptides, arginine-rich peptides, lysine-rich peptides, Pep-1 , L-oligomers, Calcitonin peptide(s), etc.
  • antimicrobial-derived CPPs e.g. Buforin-2, Bac71 5-24, SynB, SynB(l
  • Any of amino acids Arg, Lys, His, Orn and Xaa may be positioned at any place of the peptide.
  • cationic peptides or proteins in the range of 7-30 amino acids are particular preferred.
  • Even more preferred peptides of this formula are oligoarginines such as e.g. Arg 7 , Argg, Arg 9 , Arg 12 His 3 Arg 9 , Arg 9 His 3 , His 3 Arg 9 His 3 , His 6 Arg 9 His 6 His 3 Arg 4 His 3 , His 6 Arg 4 His 6 , TyrSer 2 Arg 9 Ser 2 Tyr, (ArgLysHis) 4 , Tyr(ArgLysHis) 2 Arg, etc.
  • the cationic or polycationic peptide or protein of the polymeric carrier when defined according to formula ⁇ (Arg)i;(Lys) m ;(His) protest;(Orn) 0 ;(Xaa) x ⁇ (formula (IV)) as shown above and which comprises or is additionally modified to comprise at least one -SH moiety, may be, without being restricted thereto, selected from subformula (IVa):
  • This embodiment may apply to situations, wherein the cationic or polycationic peptide or protein of the polymeric carrier, e.g. when defined according to empirical formula (Arg)i;(Lys) m ;(His) n ;(Orn) 0 ;(Xaa) x (formula (IV)) as shown above, comprises or has been modified with at least one cysteine as -SH moiety in the above meaning such that the cationic or polycationic peptide as cationic component carries at least one cysteine, which is capable to form a disulfide bond with other components of the polymeric carrier.
  • the cationic or polycationic peptide or protein of the polymeric carrier e.g. when defined according to empirical formula (Arg)i;(Lys) m ;(His) n ;(Orn) 0 ;(Xaa) x (formula (IV)) as shown above, comprises or has been modified with at least one cysteine as -SH
  • the cationic or polycationic peptide or protein of the polymeric carrier when defined according to formula ⁇ (Arg),;(Lys) m ;(His) n ;(Orn) 0 ;(Xaa) x ⁇ (formula (IV)) as shown above, may be, without being restricted thereto, selected from subformula (IVb):
  • This embodiment may apply to situations, wherein the cationic or polycationic peptide or protein of the polymeric carrier, which may be used to complex the at least one RNA of the inventive combination vaccine, e.g. when defined according to empirical formula (Arg),;(Lys) m ;(His) n ;(Orn) 0 ;(Xaa) x (formula (IV)) as shown above, has been modified with at least two cysteines as -SH moieties in the above meaning such that the cationic or polycationic peptide of the inventive polymeric carrier carries at least two (terminal) cysteines, which are capable to form a disulfide bond with other components of the polymeric carrier.
  • the cationic or polycationic peptide or protein of the polymeric carrier which may be used to complex the at least one RNA of the inventive combination vaccine, e.g. when defined according to empirical formula (Arg),;(Lys) m ;(His) n ;(Orn)
  • At least one cationic (or polycationic) component of the polymeric carrier may be selected from e.g. any (non-peptidic) cationic or polycationic polymer suitable in this context, provided that this (non-peptidic) cationic or polycationic polymer exhibits or is modified to exhibit at least one -SH-moiety, which provide for a disulfide bond linking the cationic or polycationic polymer with another component of the polymeric carrier as defined herein.
  • the polymeric carrier may comprise the same or different cationic or polycationic polymers.
  • the cationic component of the polymeric carrier comprises a (non- peptidic) cationic or polycationic polymer
  • the cationic properties of the (non-peptidic) cationic or polycationic polymer may be determined upon its content of cationic charges when compared to the overall charges of the components of the cationic polymer.
  • the content of cationic charges in the cationic polymer at a (physiological) pH as defined herein is at least 10%, 20%, or 30%, preferably at least 40%, more preferably at least 50%, 60% or 70%, but also preferably at least 80%, 90%, or even 95%, 96%, 97%, 98%, 99% or 100%, most preferably at least 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, or may be in the range of about 10% to 90%, more preferably in the range of about 30% to 100%, even preferably in the range of about 50% to 100%, e.g.
  • the (non-peptidic) cationic component of the polymeric carrier represents a cationic or polycationic polymer, typically exhibiting a molecular weight of about 0.1 or 0.5 kDa to about 100 kDa, preferably of about 1 kDa to about 75 kDa, more preferably of about 5 kDa to about 50 kDa, even more preferably of about 5 kDa to about 30 kDa, or a molecular weight of about 10 kDa to about 50 kDa, even more preferably of about 10 kDa to about 30 kDa.
  • non-peptidic cationic or polycationic polymer typically exhibits at least one -SH-moiety, which is capable to form a disulfide linkage upon condensation with either other cationic components or other components of the polymeric carrier as defined herein.
  • the (non-peptidic) cationic component of the polymeric carrier which may be used to complex the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine may be selected from acrylates, modified acrylates, such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)), chitosanes, aziridines or 2-ethyl-2-oxazoline (forming oligo ethylenimines or modified oligoethylenimines), polymers obtained by reaction of bisacrylates with amines forming oligo beta aminoesters or poly amido amines, or other polymers like polyesters, polycarbonates, etc.
  • modified acrylates such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)
  • chitosanes such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)
  • Each molecule of these (non-peptidic) cationic or polycationic polymers typically exhibits at least one -SH-moiety, wherein these at least one -SH-moiety may be introduced into the (non-peptidic) cationic or polycationic polymer by chemical modifications, e.g. using imonothiolan, 3-thio propionic acid or introduction of -SH- moieties containing amino acids, such as cysteine or any further (modified) amino acid.
  • -SH-moieties are preferably as already defined above.
  • the further component which may be contained in the polymeric carrier, and which may be used to complex the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine or which may be used to modify the different (short) cationic or polycationic peptides or (non-peptidic) polymers forming basis for the polymeric carrier or the biophysical/biochemical properties of the polymeric carrier as defined herein, is an amino acid component (AA).
  • AA amino acid component
  • the amino acid component (AA) comprises a number of amino acids preferably in a range of about 1 to 1 00, preferably in a range of about 1 to 50, more preferably selected from a number comprising 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 1 3, 14 or 1 5-20, or may be selected from a range formed by any two of the afore mentioned values.
  • the amino acids of amino acid component (AA) can be chosen independently from each other. For example if in the polymeric carrier two or more (AA) components are present they can be the same or can be different from each other.
  • the amino acid component (AA) may contain or may be flanked (e.g.
  • a -SH containing moiety which allows introducing this component (AA) via a disulfide bond into the polymeric carrier as defined herein.
  • the amino acid component (AA) may also be read as -Cys-(AA)-Cys- wherein Cys represents cysteine and provides for the necessary -SH-moiety for a disulfide bond.
  • the -SH containing moiety may be also introduced into amino acid component (AA) using any of modifications or reactions as shown above for the cationic component or any of its components.
  • the amino acid component (AA) may be provided with two -SH-moieties (or even more), e.g.
  • HS-(AA)-SH to allow binding to two functionalities via disulfide bonds, e.g. if the amino acid component (AA) is used as a linker between two further components (e.g. as a linker between two cationic polymers).
  • amino acid component (AA) may be provided with other functionalities as already described above for the other components of the polymeric carrier, which allow binding of the amino acid component (AA) to any of components of the polymeric carrier.
  • the amino acid component (AA) of the polymeric carrier may be bound to further components of the polymeric carrier, which may be used to complex the at least one NA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine with or without using a disulfide linkage.
  • the amino acid component (AA) may be used to modify the polymeric carrier, particularly the content of cationic components in the polymeric carrier as defined above.
  • the amino acid component (AA) may be selected from the following alternatives: an aromatic amino acid component, a hydrophilic (and preferably non charged polar) amino acid component, a lipophilic amino acid component, or a weak basic amino acid component.
  • the amino acid component (AA) may be a signal peptide or signal sequence, a localisation signal or sequence, a nuclear localisation signal or sequence (NLS), an antibody, a cell penetrating peptide (e.g. TAT), etc.
  • the amino acid component (AA) may be a functional peptide or protein, which may modulate the functionality of the polymeric carrier accordingly.
  • Such functional peptides or proteins as the amino acid component (AA) preferably comprise any peptides or proteins as defined herein, e.g. as defined below as therapeutically active proteins.
  • such further functional peptides or proteins may comprise so called cell penetrating peptides (CPPs) or cationic peptides for transportation.
  • CPPs cell penetrating peptides
  • the amino acid component (AA) may consist of any peptide or protein which can execute any favourable function in the cell.
  • peptides or proteins selected from therapeutically active proteins or peptides, from antigens, e.g. tumour antigens, pathogenic antigens (animal antigens, viral antigens, protozoan antigens, bacterial antigens, allergic antigens), autoimmune antigens, or further antigens, from allergens, from antibodies, from immunostimulatory proteins or peptides, from antigen- specific T cell receptors, or from any other protein or peptide suitable for a specific (therapeutic) application.
  • antigens e.g. tumour antigens, pathogenic antigens (animal antigens, viral antigens, protozoan antigens, bacterial antigens, allergic antigens), autoimmune antigens, or further antigens, from allergens, from antibodies, from immunostimulatory proteins or peptides, from antigen- specific T cell receptors, or
  • peptide epitopes from antigens particularly of the virus family Paramyxoviridae and/or of the virus family Orthomyxoviridae as defined herein, and more particularly of the F protein of the virus family Paramyxoviridae and/or of the HA protein of the virus family Orthomyxoviridae as defined herein.
  • the polymeric carrier which may be used to complex the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in inventive combination vaccine may comprise at least one of the above mentioned cationic or polycationic peptides, proteins or polymers or further components, e.g. (AA), wherein any of the above alternatives may be combined with each other, and may be formed by polymerizing same in a polymerization condensation reaction via their -SH-moieties.
  • the polymeric carrier may be selected from a polymeric carrier molecule according to generic formula (V):
  • P 1 and P 3 are different or identical to each other and represent a linear or branched hydrophilic polymer chain, each P 1 and P 3 exhibiting at least one -SH-moiety, capable to form a disulfide linkage upon condensation with component P 2 , or alternatively with (AA), (AA) X , or [(AA)J Z if such components are used as a linker between P 1 and P 2 or P 3 and P 2 ) and/or with further components (e.g.
  • the linear or branched hydrophilic polymer chain selected independent from each other from polyethylene glycol (PEG), poly-/V-(2- hydroxypropyl)methacrylamide, poly-2-(methacryloyloxy)ethyl phosphorylcholines, poly(hydroxyalkyl L-asparagine), poly(2- (methacryloyloxy)ethyl phosphorylcholine), h y d roxyeth y I sta rc h or poly(hydroxyalkyl L-glutamine), wherein the hydrophilic polymer chain exhibits a molecular weight of about 1 kDa to about 100 kDa, preferably of about 2 kDa to about 25 kDa; or more preferably of about 2 kDa to about 1 0 kDa, e.g.
  • a cationic or polycationic peptide or protein is a cationic or polycationic peptide or protein, e.g. as defined above for the polymeric carrier formed by disulfide-crosslinked cationic components, and preferably having a length of about 3 to about 1 00 amino acids, more preferably having a length of about 3 to about 50 amino acids, even more preferably having a length of about 3 to about 25 amino acids, e.g.
  • the polymeric carrier formed by disulfide-crosslinked cationic components typically having a molecular weight of about 0.5 kDa to about 30 kDa, including a molecular weight of about 1 kDa to about 20 kDa, even more preferably of about 1 .5 kDa to about 10 kDa, or having a molecular weight of about 0.5 kDa to about 100 kDa, including a molecular weight of about 10 kDa to about 50 kDa, even more preferably of about 10 kDa to about 30 kDa; each P 2 exhibiting at least two -SH-moieties, capable to form a disulfide linkage upon condensation with further components P 2 or component(s) P' and/or P 3 or alternatively with further components (e.g.
  • (AA), (AA) X , or [(AA)J Z ); is a (reversible) disulfide bond (the brackets are omitted for better readability), wherein S preferably represents sulphur or a -SH carrying moiety, which has formed a (reversible) disulfide bond.
  • the (reversible) disulfide bond is preferably formed by condensation of -SH-moieties of either components P 1 and P 2 , P 2 and P 2 , or P 2 and P 3 , or optionally of further components as defined herein (e.g.
  • the -SH-moiety may be part of the structure of these components or added by a modification as defined below; is an optional ligand, which may be present or not, and may be selected independent from the other from RGD, Transferrin, Folate, a signal peptide or signal sequence, a localization signal or sequence, a nuclear localization signal or sequence (NLS), an antibody, a cell penetrating peptide, (e.g. TAT or KALA), a ligand of a receptor (e.g. cytokines, hormones, growth factors etc), small molecules (e.g.
  • n is an integer, typically selected from a range of about 1 to 50, preferably from a range of about 1 , 2 or 3 to 30, more preferably from a range of about 1 , 2, 3, 4, or 5 to 25, or a range of about 1 , 2, 3, 4, or 5 to 20, or a range of about 1 , 2, 3, 4, or 5 to 15, or a range of about 1 , 2, 3, 4, or 5 to 10, including e.g.
  • n is in a range of about 1 , 2, 3, 4, or 5 to 10, more preferably in a range of about 1 , 2, 3, or 4 to 9, in a range of about 1 , 2, 3, or 4 to 8, or in a range of about 1 , 2, or 3 to 7.
  • Each of hydrophilic polymers P 1 and P 3 typically exhibits at least one -SH-moiety, wherein the at least one -SH-moiety is capable to form a disulfide linkage upon reaction with component P 2 or with component (AA) or (AA) X , if used as linker between P 1 and P 2 or P 3 and P 2 as defined below and optionally with a further component, e.g. L and/or (AA) or (AA) X , e.g. if two or more -SH-moieties are contained.
  • the term "-S-S-" in these formulae may also be written as “-S-Cys", as “-Cys-S” or as “-Cys-Cys-”.
  • the term “-Cys-Cys-” does not represent a peptide bond but a linkage of two cysteines via their -SH-moieties to form a disulfide bond.
  • the term “-Cys-Cys-” also may be understood generally as “-(Cys-S)-(S-Cys)-", wherein in this specific case S indicates the sulphur of the -SH-moiety of cysteine.
  • the terms "-S- Cys” and “-Cys-S” indicate a disulfide bond between a -SH containing moiety and a cysteine, which may also be written as “-S-(S-Cys)" and "-(Cys-S)-S".
  • the hydrophilic polymers P 1 and P 3 may be modified with a -SH moiety, preferably via a chemical reaction with a compound carrying a -SH moiety, such that each of the hydrophilic polymers P' and P 3 carries at least one such -SH moiety.
  • a compound carrying a -SH moiety may be e.g.
  • Such a compound may also be any non-amino compound or moiety, which contains or allows to introduce a -SH moiety into hydrophilic polymers P 1 and P 3 as defined herein.
  • Such non-amino compounds may be attached to the hydrophilic polymers P 1 and P 3 of formula (VI) of the polymeric carrier according to the present invention via chemical reactions or binding of compounds, e.g. by binding of a 3-thio propionic acid or thioimolane, by amide formation (e.g.
  • alkenes or alkines imine or hydrozone formation (aldehydes or ketons, hydrazins, hydroxylamins, amines), complexation reactions (avidin, biotin, protein G) or components which allow S n - type substitution reactions (e.g halogenalkans, thiols, alcohols, amines, hydrazines, hydrazides, sulphonic acid esters, oxyphosphonium salts) or other chemical moieties which can be utilized in the attachment of further components.
  • a particularly preferred PEG derivate in this context is alpha-Methoxy-omega-mercapto poly(ethylene glycol).
  • the SH-moiety e.g.
  • each of hydrophilic polymers P 1 and P 3 typically exhibits at least one -SH-moiety preferably at one terminal end, but may also contain two or even more - SH-moieties, which may be used to additionally attach further components as defined herein, preferably further functional peptides or proteins e.g. a ligand, an amino acid component (AA) or (AA) X , antibodies, cell penetrating peptides or enhancer peptides (e.g. TAT, KALA), etc.
  • further functional peptides or proteins e.g. a ligand, an amino acid component (AA) or (AA) X , antibodies, cell penetrating peptides or enhancer peptides (e.g. TAT, KALA), etc.
  • the amino acid component (AA) or (AA) X in the polymeric carrier of formula (V or VI) may also occur as a mixed repetitive amino acid component [(AA)J Z , wherein the number of amino acid components (AA) or (AA) X is further defined by integer z.
  • z may be selected from a range of about 1 to 30, preferably from a range of about 1 to 15, more preferably 1 to 10 or 1 to 5 and even more preferably selected from a number selected from 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14 or 15, or may be selected from a range formed by any two of the afore mentioned values.
  • the amino acid component (AA) or (AA) X may be used to modify component P 2 , particularly the content of component S-P 2 -S in repetitive component [S-P 2 - S] n of the polymeric carrier of formula (V) above.
  • This may be represented in the context of the entire polymeric carrier according to formula (VI) e.g. by following formula (Via):
  • any of the single components [S-P 2 -S] and [S-(AA) X -S] may occur in any order in the subformula ⁇ [S-P 2 -S] a [S-(AA) x -S] b ⁇ .
  • the polymeric carrier which may be used to complex the at least one RNA of the combination vaccine or any further nucleic acid comprised in the combination vaccine or single components thereof, e.g. of the above mentioned cationic or polycationic peptides, proteins or polymers or further components, e.g. (AA), may be further modified with a ligand, preferably a carbohydrate, more preferably a sugar, even more preferably mannose.
  • a ligand preferably a carbohydrate, more preferably a sugar, even more preferably mannose.
  • the entire polymeric carrier may be formed by a polymerization condensation (of at least one) of the above mentioned cationic or polycationic peptides, proteins or polymers or further components, e.g. (AA), via their -SH- moieties in a first step and complexing the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine to such a polymeric carrier in a second step.
  • the polymeric carrier may thus contain a number of at least one or even more of the same or different of the above defined cationic or polycationic peptides, proteins or polymers or further components, e.g. (AA), the number preferably determined by the above range.
  • the polymeric carrier which may be used to complex the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine is formed by carrying out the polymerization condensation of at least one of the above mentioned cationic or polycationic peptides, proteins or polymers or further components, e.g. (AA), via their -SH-moieties simultaneously to complexing the at least one RNA of the inventive combination vaccine or any further nucleic acid comprised in the inventive combination vaccine to the (in situ prepared) polymeric carrier.
  • AA cationic or polycationic peptides, proteins or polymers or further components
  • the polymeric carrier may thus also here contain a number of at least one or even more of the same or different of the above defined cationic or polycationic peptides, proteins or polymers or further components, e.g. (AA), the number preferably determined by the above range.
  • AA cationic or polycationic peptides, proteins or polymers or further components
  • the at least one RNA of the inventive combination vaccine or any further coding nucleic acid in the inventive combination vaccine is complexed at least partially with a cationic or polycationic compound and/or a polymeric carrier, preferably cationic proteins or peptides.
  • a cationic or polycationic compound and/or a polymeric carrier, preferably cationic proteins or peptides.
  • the ratio of complexed RNA to: free RNA in the combination vaccine is selected from a range, of about 5:1 (w/w) to about 1 :10 (w/w), more preferably from a range of about 4:1 (w/w) to about 1 :8 (w/w), even more preferably from a range of about 3:1 (w/w) to about 1 :5 (w/w) or 1 :3 (w/w), and most preferably the ratio of complexed RNA to free RNA in the inventive combination vaccine is selected from a ratio of about 1 :1 (w/w).
  • the complexed RNA in the inventive combination vaccine is preferably prepared according to a first step by complexing the at least one RNA with a cationic or polycationic compound and/or with a polymeric carrier, preferably as defined herein, in a specific ratio to form a stable complex.
  • a cationic or polycationic compound or polymeric carrier preferably as defined herein, in a specific ratio to form a stable complex.
  • the ratio of the RNA and the cationic or polycationic compound and/or the polymeric carrier in the component of the complexed RNA is typically selected in a range that the RNA is entirely complexed and no free cationic or polycationic compound or polymeric carrier or only a negligibly smal l amount thereof remains in the composition.
  • the ratio of the RNA e.g.
  • mRNA) to the cationic or polycationic compound and/or the polymeric carrier is selected from a range of about 6:1 (w/w) to about 0,25:1 (w/w), more preferably from about 5: 1 (w/w) to about 0,5:1 (w/w), even more preferably of about 4: 1 (w/w) to about 1 :1 (w/w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ratio of about 3:1 (w/w) to about 2:1 (w/w).
  • the ratio of the RNA to the cationic or polycationic compound and/or the polymeric carrier, preferably as defined herein, in the component of the complexed RNA may also be calculated on the basis of the nitrogen/phosphate ratio (N/P-ratio) of the entire complex.
  • an N/P-ratio is preferably in the range of about 0.1 -1 0, preferably in a range of about 0.3-4 and most preferably in a range of about 0.5-2 or 0.7-2 regarding the ratio of RNA: cationic or polycationic compound and/or polymeric carrier, preferably as defined herein, in the complex, and most preferably in a range of about 0.7- 1 ,5, 0.5-1 or 0.7-1 , and even most preferably in a range of about 0.3-0.9 or 0.5-0.9., preferably provided that the cationic or polycationic compound in the complex is a cationic or polycationic cationic or polycationic protein or peptide and/or the polymeric carrier as defined above.
  • the complexed RNA is also emcompassed in the term "adjuvant component".
  • the at least one antigen-providing RNA of the inventive combination vaccine as defined above may be formulated together with an adjuvant.
  • an adjuvant may be preferably a further nucleic acid that is not encoding a further antigen but is able to stimulate an unspecific immune response, i.e. innate immune response, by interacting with any part of the innate immune system.
  • an adjuvant nucleic acid Such a nucleic acid stimulating an unspecific immune response is termed herein as "adjuvant nucleic acid”.
  • an adjuvant nucleic acid preferably comprises or consists of an oligo- or a polynucleotide; more preferably an adjuvant nucleic acid comprises or consists of a RNA or a DNA; even more preferably such an adjuvant nucleic acid comprising or consisting of a RNA or a DNA being complexed with a cationic or polycationic compound and/or with a polymeric carrier; optionally in a weight ratio selected from a range of about 6:1 (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/w) to about 0.5: 1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w:w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ration of about 3:1 (w/w) to about 2:1 (w/w) of adjuvant component to cationic or polycationic compound and/or with a
  • a combination vaccine according to the invention may comprise a first RNA encoding for an antigenic function, e.g. the RSV Fusion (F) protein, the Influenza Hemagglutinin (HA) protein or the like, and a second nucleic acid that is acting as an adjuvant which is called the adjuvant nucleic acid.
  • the inventive combination vaccine may also comprise further RNAs encoding for further antigenic functions and is also not limited to comprise only one adjuvant nucleic acid but can comprise several different of them.
  • Both kinds of nucleic acid, the antigen-encoding RNA and the adjuvant nucleic acid may be, independently from each other, complexed with a carrier as defined above. Therefore, a cationic or polycationic compound and/or a polymeric carrier used to complex the at least one adjuvant nucleic acid, may be selected from a cationic or polycationic compound and/or a polymeric carrier as defined above.
  • an adjuvant nucleic acid is preferably selected from nucleic acids which are known to bind to TLR receptors.
  • Such an adjuvant nucleic acid can be in the form of a(n) (immunostimulatory) CpG nucleic acid, in particular CpG-RNA or CpG- DNA, which preferably induces an innate immune response.
  • a CpG-RNA or CpG-DNA used according to the invention can be a single-stranded CpG-DNA (ss CpG-DNA), a double-stranded CpG-DNA (dsDNA), a single-stranded CpG-RNA (ss CpG-RNA) or a double-stranded CpG-RNA (ds CpG-RNA).
  • the CpG nucleic acid used according to the invention is preferably in the form of CpG-RNA, more preferably in the form of single- stranded CpG-RNA (ss CpG-RNA).
  • such CpG nucleic acids have a length as described above.
  • the CpG motifs are unmethylated.
  • an adjuvant nucleic acid can be an immunostimulatory RNA (isRNA), which preferably elicits an innate immune response.
  • the immunostimulatory RNA may be a single-stranded, a double-stranded or a partially double- stranded RNA, more preferably a single-stranded RNA, and/or a circular or linear RNA, more preferably a linear RNA.
  • the immunostimulatory RNA may be a (linear) single-stranded RNA.
  • the immunostimulatory RNA may be a (long) (linear) (single-stranded) non-coding RNA.
  • the isRNA carries a triphosphate at its 5'-end which is the case for in vitro transcribed RNA.
  • An immunostimulatory RNA may also occur as a short RNA oligonucleotide as defined herein.
  • An immunostimulatory RNA as used herein may furthermore be selected from any class of RNA molecules, found in nature or being prepared synthetically, and which can induce an innate immune response and may support an adaptive immune response induced by an antigen.
  • the inventive combination vaccine comprises an antigen-providing RNA and additionally an adjuvant nucleic acid
  • the immune response that is evoked by administration of such a vaccine comprises activation of both parts of the immune system, the adaptive immune system as well as the innate immune system.
  • T-lymphocytes are typically divided into two sub-populations, the T- helper 1 cells, in the following Thl -cells, and the T-helper 2 cells, in the following Th2-cells, with which the immune system is capable of destroying intracellular and extracellular pathogens (e.g. antigens).
  • Th1 -cells are responsible for intracellular pathogen destruction by assisting the cellular immune response by activation of macrophages and cytotoxic T cells.
  • Th2-cells are mainly for extracellular pathogen- elimination and promote the humoral immune response by stimulation of B-cells for conversion into plasma cells and by formation of antibodies (e.g.
  • the two T-helper cell populations differ in the pattern of the effector proteins (cytokines) produced by them.
  • the Th1 -cell/Th2-cell ratio is of great importance in the induction and maintenance of an adaptive immune response.
  • the Th1 -cell Th2-cell ratio of the (adaptive) immune response is preferably shifted in the direction towards the cellular response (Th1 response) and a cellular immune response is thereby induced. Stimulation of this response of the adaptive immune system is mainly provoked by the translation of the antigen-providing RNA and the resulting presence of the peptide or protein antigens within the organism.
  • TLRs Toll-like receptors
  • PRR pattern recognition receptor
  • PAMPs pathogen-associated molecular patterns
  • TLR1 - TLR13 Toll-like receptors: TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR1 1 , TLR12 or TLR1 3
  • TLR1 - TLR13 Toll-like receptors: TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR1 1 , TLR12 or TLR1 3
  • ligands for TLR9 include certain nucleic acid molecules and that certain types of RNA are immunostimulatory in a sequence-independent or sequence-dependent manner, wherein these various immunostimulatory RNAs may e.g. stimulate TLR3, TLR7, or TLR8, or intracellular receptors such as RIG-I, MDA-5, etc.
  • an adjuvant nucleic acid preferably an immunostimulatory RNA (isRNA), as used herein, may comprise any RNA sequence known to be immunostimulatory, including, e.g., RNA sequences representing and/or encoding ligands of TLRs, preferably selected from human family members TLR1 - TLR10 or murine family members TLR1 - TLR1 3, more preferably selected from (human) family members TLR1 - TLRT0, even more preferably from TLR7 and TLR8, ligands for intracellular receptors for RNA (such as RIG-I or MDA-5, etc.) (see e.g.
  • immunostimulatory RNA molecules used as a further compound of the inventive combination vaccine, may include any other RNA capable of eliciting an innate immune response.
  • an immunostimulatory RNA may include ribosomal RNA (rRNA), transfer RNA (tRNA), messenger RNA (mRNA), and viral RNA (vRNA).
  • an immunostimulatory RNA may comprise a length of 1000 to 5000, of 500 to 5000, of 5 to 5000, or of 5 to 1000, 5 to 500, 5 to 250, of 5 to 100, of 5 to 50 or of 5 to 30 nucleotides.
  • an adjuvant nucleic acid sequence, particularly an isRNA, as used herein may consist of or comprise a nucleic acid of formula (VII) or (VIII):
  • G is guanosine, uracil or an analogue of guanosine or uracil;
  • X is guanosine, uracil, adenosine, thymidine, cytosine or an analogue of the above- mentioned nucleotides
  • I is an integer from 1 to 40
  • n is an integer from 1 to 40
  • n > 1 at least 50% of the nucleotides are guanosine or an analogue thereof.
  • C is cytosine, uracil or an analogue of cytosine or uracil;
  • X is guanosine, uracil, adenosine, thymidine, cytosine or an analogue of the above- mentioned nucleotides
  • I is an integer from 1 to 40
  • n is an integer from 1 to 40
  • n > 1 at least 50% of the nucleotides are cytosine or an analogue thereof.
  • nucleic acids of formula (VII) or (VIII), which may be used as an adjuvant nucleic acid sequence, particularly an isRNA may be relatively short nucleic acid molecules with a typical length of approximately from 5 to 100 (but may also be longer than 100 nucleotides for specific embodiments, e.g. up to 200 nucleotides), from 5 to 90 or from 5 to 80 nucleotides, preferably a length of approximately from 5 to 70, more preferably a length of approximately from 8 to 60 and, more preferably a length of approximately from 15 to 60 nucleotides, more preferably from 20 to 60, most preferably from 30 to 60 nucleotides.
  • the number of nucleotides G in the nucleic acid of formula (I) is determined by I or n.
  • a nucleotide adjacent to X m in the nucleic acid of formula (VII) according to the invention is preferably not a uracil.
  • the number of nucleotides C in the nucleic acid of formula (VIII) according to the invention is determined by I or n.
  • a nucleotide adjacent to X m in the nucleic acid of formula (VIII) according to the invention is preferably not a uracil.
  • I or n > 1 at least 60%, 70%, 80%, 90% or even 100% of the nucleotides are guanosine or an analogue thereof, as defined above.
  • the remaining nucleotides to 100% (when guanosine constitutes less than 100% of the nucleotides) in the flanking sequences G, and/or G n are uracil or an analogue thereof, as defined hereinbefore.
  • I and n independently of one another, are each an integer from 2 to 30, more preferably an integer from 2 to 20 and yet more preferably an integer from 2 to 1 5.
  • the lower limit of I or n can be varied if necessary and is at least 1 , preferably at least 2, more preferably at least 3, 4, 5, 6, 7, 8, 9 or 10. This definition applies correspondingly to formula (VIII).
  • an immunostimulatory nucleic acid sequence may consist of or comprise a nucleic acid of formula (IX) or (X):
  • G is guanosine (guanine), uridine (uracil) or an analogue of guanosine (guanine) or uridine (uracil), preferably guanosine (guanine) or an analogue thereof;
  • X is guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine), or an analogue of these nucleotides (nucleosides), preferably uridine (uracil) or an analogue thereof;
  • N is a nucleic acid sequence having a length of about 4 to 50, preferably of about 4 to 40, more preferably of about 4 to 30 or 4 to 20 nucleic acids, each N independently being selected from guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of these nucleotides
  • a is an integer from 1 to 20, preferably from 1 to 1 5, most preferably from 1 to 10;
  • I is an integer from 1 to 40,
  • G is guanosine (guanine) or an analogue thereof
  • nucleosides when I > 1 , at least 50% of these nucleotides (nucleosides) are guanosine (guanine) or an analogue thereof;
  • n is an integer and is at least 3;
  • X is uridine (uracil) or an analogue thereof
  • n is an integer from 1 to 40
  • G is guanosine (guanine) or an analogue thereof, when n > 1 , at least 50% of these nucleotides (nucleosides) are guanosine (guanine) or an analogue thereof;
  • u, v may be independently from each other an integer from 0 to 50,
  • nucleic acid molecule of formula (IX) has a length of at least 50 nucleotides, preferably of at least 100 nucleotides, more preferably of at least 150 nucleotides, even more preferably of at least 200 nucleotides and most preferably of at least 250 nucleotides.
  • C is cytidine (cytosine), uridine (uracil) or an analogue of cytidine (cytosine) or uridine
  • uracil preferably cytidine (cytosine) or an analogue thereof;
  • X is guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of the above-mentioned nucleotides (nucleosides), preferably uridine (uracil) or an analogue thereof;
  • N is each a nucleic acid sequence having independent from each other a length of about 4 to 50, preferably of about 4 to 40, more preferably of about 4 to 30 or 4 to
  • nucleic acids each N independently being selected from guanosine (guanine), uridine (uracil), adenosine (adenine), thymidine (thymine), cytidine (cytosine) or an analogue of these nucleotides (nucleosides);
  • a is an integer from 1 to 20, preferably from 1 to 1 5, most preferably from 1 to 10;
  • I is an integer from 1 to 40,
  • C is cytidine (cytosine) or an analogue thereof
  • nucleosides when I > 1 , at least 50% of these nucleotides (nucleosides) are cytidine (cytosine) or an analogue thereof;
  • n is an integer and is at least 3;
  • X is uridine (uracil) or an analogue thereof
  • n is an integer from 1 to 40
  • C is cytidine (cytosine) or an analogue thereof
  • nucleosides when n > 1 , at least 50% of these nucleotides (nucleosides) are cytidine (cytosine) or an analogue thereof,
  • u, v may be independently from each other an integer from 0 to 50,
  • nucleic acid molecule of formula (X) has a length of at least 50 nucleotides, preferably of at least 100 nucleotides, more preferably of at least 1 50 nucleotides, even more preferably of at least 200 nucleotides and most preferably of at least 250 nucleotides.
  • the definition of bordering elements N u and N v in formula (X) is identical to the definitions given above for N u and N v in formula (IX).
  • the adjuvant which may be used together with the antigen-providing RNA in the inventive combination vaccine, is preferably prepared according to a first step by complexing the immunostimulatory RNA (isRNA) with a cationic or polycationic compound and/or with a polymeric carrier, preferably as defined herein, in a specific ratio to form a stable complex.
  • a cationic or polycationic compound or polymeric carrier preferably as defined herein, in a specific ratio to form a stable complex.
  • it is highly preferable that no free cationic or polycationic compound or polymeric carrier or only a negligibly small amount thereof remains in the adjuvant after complexing the isRNA.
  • the ratio of the isRNA and the cationic or polycationic compound and/or the polymeric carrier in the adjuvant is typically selected in a range that the isRNA is entirely complexed and no free cationic or polycationic compound or polymeric carrier or only a negligibly small amount thereof remains in the composition.
  • the ratio of the adjuvant i.e. the ratio of the isRNA to the cationic or polycationic compound and/or the polymeric carrier, preferably as defined herein, is selected from a range of about 6:1 (w/w) to about 0,25:1 (w/w), more preferably from about 5:1 (w/w) to about 0,5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w/w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ratio of about 3:1 (w/w) to about 2:1 (w/w).
  • the ratio of the isRNA to the cationic or polycationic compound and/or the polymeric carrier, preferably as defined herein, in the adjuvant may also be calculated on the basis of the nitrogen/phosphate ratio (N/P-ratio) of the entire complex of the adjuvant.
  • an N/P-ratio is preferably in the range of about 0.1 - 10, preferably in a range of about 0.3-4 and most preferably in a range of about 0.5-2 or 0.7- 2 regarding the ratio of isRNA: cationic or polycationic compound and/or polymeric carrier, preferably as defined herein, in the complex, and most preferably in a range of about 0.7- 1 ,5, 0.7-1 or 0.5-1, and even most preferably in a range of about 0.3-0.9 or 0.5-0.9., preferably provided the cationic or polycationic compound in the complex is a cationic or polycationic cationic or polycationic protein or peptide and/or the polymeric carrier is as defined above.
  • the inventive combination vaccine is formulated to comprise a) said at least one RNA; preferably in form of a mono-, bi- or multicistronic RNA, optionally being stabilized, optionally being optimized for translation and/or optionally being complexed with a cationic or polycationic compound or a polymeric carrier;
  • an adjuvant component comprising or consisting of said at least one RNA and/or at least one adjuvant nucleic acid, complexed with a cationic or polycationic compound and/or with a polymeric carrier, and
  • the optionally comprised adjuvant component comprises the same RNA as comprised in the inventive combination vaccine as antigen- providing RNA e.g. mRNA coding for a F protein of viruses of the Paramyxoviridae or fragments, variants or derivatives thereof, or coding for a HA protein of viruses of the Orthomyxoviridae or fragments, variants or derivatives thereof.
  • inventive combination vaccine may comprise further components for facilitating administration and uptake of the vaccine. Such further components may be an appropriate carrier or vehicle, additional adjuvants for supporting any immune response, antibacterial and/or antiviral agents.
  • the inventive combination vaccine furthermore comprises a pharmaceutically acceptable carrier and/or vehicle.
  • a pharmaceutically acceptable carrier typically includes the liquid or non-liquid basis of a composition comprising the components of the combination vaccine. If the composition is provided in liquid form, the carrier will typically be pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g. phosphate, citrate etc. buffered solutions.
  • the injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e.
  • the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects.
  • Reference media are e.g. liquids occurring in "in vivd' methods, such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in "in vitrd' methods, such as common buffers or liquids. Such common buffers or liquids are known to a skilled person. Ringer-Lactate solution is particularly preferred as a liquid basis.
  • compatible solid or liquid fillers or diluents or encapsulating compounds which are suitable for administration to a patient to be treated, may be used as well for the vaccine according to the invention.
  • compatible means that these constituents of the combination vaccine are capable of being mixed with the components of the combination vaccine in such a manner that no interaction occurs which would substantially reduce the pharmaceutical effectiveness of the combination vaccine under typical use conditions.
  • the inventive combination vaccine may comprise one or more additional adjuvants which are suitable to initiate or increase an immune response of the innate immune system, i.e. a non-specific immune response, particularly by binding to pathogen- associated molecular patterns (PAMPs).
  • PAMPs pathogen- associated molecular patterns
  • the vaccine when administered, the vaccine preferably elicits an innate immune response due to the adjuvant, optionally contained therein.
  • an adjuvant may be selected from an adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal, e.g. an adjuvant protein as defined above or an adjuvant as defined in the following.
  • an adjuvant may be selected from an adjuvant as defined above.
  • an adjuvant may be selected from any adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an innate immune response in a mammal and/or suitable for depot and delivery of the components of the combination vaccine.
  • adjuvants suitable for depot and delivery are cationic or polycationic compounds as defined above.
  • the adjuvant may be selected from the group consisting of, e.g., cationic or polycationic compounds as defined above, from chitosan, TDM, MDP, muramyl dipeptide, pluronics, alum solution, aluminium hydroxide, ADJUMERTM (polyphosphazene); aluminium phosphate gel; glucans from algae; algammulin; aluminium hydroxide gel (alum); highly protein-adsorbing aluminium hydroxide gel; low viscosity aluminium hydroxide gel; AF or SPT (emulsion of squalane (5%), Tween 80 (0.2%), Pluronic L121 (1.25%), phosphate-buffered saline, pH 7.4); AVRIDINETM (propanediamine); BAY R1005TM ((N-(2-deoxy-2-L-leucylaminob- D- glucopyranosyl)-N-octadecyl-dode
  • TM liposomes
  • LOXORIBINETM (7-allyl-8-oxoguanosine); LT 5 oral adjuvant (Eco// labile enterotoxin-protoxin); microspheres and microparticles of any composition; MF59TM; (squalenewater emulsion); MONTANIDE ISA 51 TM (purified incomplete Freund's adjuvant); MONTANIDE ISA 720TM (metabolisable oil adjuvant); MPLTM (3-Q-desacyl-4'-monophosphoryl lipid A); MTP-PE and MTP-PE liposomes ((N- acetyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(l,2-dipalmitoyl-sn-glycero-3-
  • MURAMETIDETM Nac-Mur-L-Ala- D-Gln-OCH3
  • MURAPALMITINETM and DMURAPALMITINETM Nac-Mur-L-Thr-D- isoGln-sn-glyceroldipalmitoyl
  • NAGO neuroaminidase- galactose oxidase
  • nanospheres or nanoparticles of any composition NISVs (non-ionic surfactant vesicles); PLEURANTM ( 3 ⁇ 4- glucan); PLGA, PGA and PLA (homo- and co-polymers of lactic acid and glycolic acid; microspheres/nanospheres); PLURONIC L121 TM; PMMA (polymethylmethacrylate); PODDSTM (proteinoid microspheres); polyethylene carbamate derivatives; poly-rA: poly-rU (polya
  • an adjuvant may be selected from adjuvants, which support induction of a Th1 -immune response or maturation of naive T-cells, such as GM-CSF, IL-12, IFNg, any immunostimulatory nucleic acid as defined above, preferably an immunostimulatory RNA, CpG DNA, etc.
  • the inventive combination vaccine contains besides the antigen-providing RNA further components which are selected from the group comprising: further antigens or further antigen-providing nucleic acids; a further immunotherapeutic agent; one or more auxiliary substances; or any further compound, which is known to be immunostimulating due to its binding affinity (as ligands) to human Toll-like receptors; and/or an adjuvant nucleic acid, preferably an immunostimulatory RNA (isRNA).
  • further components which are selected from the group comprising: further antigens or further antigen-providing nucleic acids; a further immunotherapeutic agent; one or more auxiliary substances; or any further compound, which is known to be immunostimulating due to its binding affinity (as ligands) to human Toll-like receptors; and/or an adjuvant nucleic acid, preferably an immunostimulatory RNA (isRNA).
  • a further component of the inventive combination vaccine may be an immunotherapeutic agent that can be selected from immunoglobulins, preferably IgGs, monoclonal or polyclonal antibodies, polyclonal serum or sera, etc, most preferably immunoglobulins directed against a virus of the Paramyxoviridae family e.g. palivizumab.
  • a further immunotherapeutic agent may be provided as a peptide/protein or may be encoded by a nucleic acid, preferably by a DNA or an RNA, more preferably an mRNA.
  • Such an immunotherapeutic agent allows providing passive vaccination additional to active vaccination triggered by the RNA encoded antigens of the inventive combination vaccine.
  • the inventive combination vaccine can additionally contain one or more auxiliary substances in order to increase its immunogenicity or immunostimulatory capacity, if desired.
  • a synergistic action of the components of the inventive combination vaccine and of an auxiliary substance, which may be optionally contained in the vaccine, is preferably achieved thereby.
  • various mechanisms can come into consideration in this respect. For example, compounds that permit the maturation of dendritic cells (DCs), for example lipopolysaccharides, TNF-alpha or CD40 ligand, form a first class of suitable auxiliary substances.
  • DCs dendritic cells
  • TNF-alpha or CD40 ligand form a first class of suitable auxiliary substances.
  • auxiliary substance any agent that influences the immune system in the manner of a "danger signal" (LPS, GP96, etc.) or cytokines, such as GM-CSF, which allow an immune response to be enhanced and/or influenced in a targeted manner.
  • a "danger signal” LPS, GP96, etc.
  • cytokines such as GM-CSF
  • auxiliary substances are cytokines, such as monokines, lymphokines, interleukins or chemokines, that further promote the innate immune response, such as IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-1 3, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21 , IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31 , IL-32, IL-33, IFN- alpha, IFN-beta, IFN-gamma, GM-CSF, G-CSF, M-CSF, LT-beta or TNF-alpha, growth factors, such as hGH.
  • cytokines such as monokines, lymphokines, inter
  • the inventive combination vaccine can also additionally contain any further compound, which is known to be immunostimulating due to its binding affinity (as ligands) to human Toll-like receptors TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLRI O, or due to its binding affinity (as ligands) to murine Toll-like receptors TLR1 , TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLRl 1 , TLR12 or TLR13, a ligand of a NOD-like receptor, or a ligand of a RIG-I like receptor.
  • the inventive combination vaccine may also additionally contain an adjuvant nucleic acid, preferably an immunostimulatory RNA (isRNA), as defined above.
  • the inventive combination vaccine furthermore comprises at least one adjuvant, an auxiliary substance selected from lipopolysaccharides, TNF-alpha, CD40 ligand, or cytokines, monokines, lymphokines, interleukins or chemokines, IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL- 13, IL-14, IL-15, IL-16, IL-1 7, IL-18, IL-19, IL-20, IL-21 , IL-22, IL-23, IL-24, IL-25, IL-26, IL- 27, IL-28, IL-29, IL-30, IL-31 , IL-32, IL-33, IF -alpha, IFN-beta, IFN-gamma, GM-CSF, G- CSF, M-CSF, LT-beta,
  • antigens additionally included in the inventive combination vaccine are typically substances such as cells, proteins, peptides, nucleic acids or other bio- or macromolecules or fragments thereof. More preferably, antigens may be proteins and peptides or fragments thereof, such as epitopes of those proteins or peptides, preferably having 5 to 1 5, more preferably 6 to 9, amino acids. Particularly, said proteins, peptides or epitopes may be derived from Hemagglutinin (HA)-proteins of viruses of the Orthomyxoviridae or may be derived from Fusion (F) proteins of viruses of the Paramyxoviridae. Further, antigens may also comprise any other biomolecule, e.g., lipids, carbohydrates, etc.
  • HA Hemagglutinin
  • F Fusion
  • the antigen is a protein or (poly-) peptide antigen, a nucleic acid, a nucleic acid encoding a protein or (poly-) peptide antigen, a polysaccharide antigen, a polysaccharide conjugate antigen, a lipid antigen, a glycolipid antigen, a carbohydrate antigen, a bacterium, a cell (vaccine), or killed or attenuated viruses or bacteria.
  • a protein or (poly-) peptide antigen a nucleic acid, a nucleic acid encoding a protein or (poly-) peptide antigen, a polysaccharide antigen, a polysaccharide conjugate antigen, a lipid antigen, a glycolipid antigen, a carbohydrate antigen, a bacterium, a cell (vaccine), or killed or attenuated viruses or bacteria.
  • antigens of the virus family Paramyxoviridae particularly the addition of antigens comprising or coding for full-length or fragments, variants or derivatives of: F- fusion protein, N -nucleocapsid protein, P - phosphoprotein, M - matrix protein, SH - small hyrdophobic protein, G -glycoprotein, NS1 - non-structural protein 1 , NS2 - non-structural protein 2, M2-1 - elongation factor, M2-2 - transcription regulation and/or L -large protein, or the addition of antigens of the virus family Orthomyxoviridae, particularly the addition of antigens comprising or coding for full-length or fragments, variants or derivatives of: HA - Hemagglutinin, NA - Neuraminidase, NP - Nucleoprotein, M1 - matrix protein 1 , M2 - matrix protein 2, NEP - nuclear export protein, PA
  • the combination vaccine as defined according to the present invention may furthermore comprise further additives or additional compounds.
  • Further additives which may be included in the combination vaccine are emulsifiers, such as, for example, Tween ® ; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet-forming agents; stabilizers; antioxidants; preservatives, RNase inhibitors and/or an anti-bacterial agent or an anti-viral agent.
  • any anti -bacterial agents known to one of skill in the art may be used in combination with the components of the inventive combination vaccine as defined herein.
  • anti-bacterial agents include Amikacin, Amoxicillin, Amoxicillin- clavulanic acid, Amphothericin-B, Ampicillin, Ampicllin-sulbactam, Apramycin, Azithromycin, Aztreonam, Bacitracin, Benzylpenicillin, Caspofungin, Cefaclor, Cefadroxil, Cefalexin, Cefalothin, Cefazolin, Cefdinir, Cefepime, Cefixime, Cefmenoxime, Cefoperazone, Cefoperazone-sulbactam, Cefotaxime, Cefoxitin, Cefbirome, Cefpodoxime, Cefpodoxime-clavulanic acid, Cefpodoxime-sulbactam, Cefbrozil, Cefquinome, Ceftazidime
  • Dalfopristin/Quinopristin Daptomycin, Dibekacin, Dicloxacillin, Doripenem, Doxycycline, Enrofloxacin, Ertapenem, Erythromycin, Flucloxacillin, Fluconazol, Flucytosin, Fosfomycin, Fusidic acid, Garenoxacin, Gatifloxacin, Gemifloxacin, Gentamicin, Imipenem, Itraconazole, Kanamycin, Ketoconazole, Levofloxacin, Li neomycin, Linezolid, Loracarbef, Mecillnam (amdinocillin), Meropenem, Metronidazole, Meziocillin, Mezlocillin- sulbactam, Minocycline, Moxifloxacin, Mupirocin, Nalidixic acid, Neomycin, Netilmicin, Nitrofurantoin, Norfloxacin, Ofloxacin, Oxacillin
  • Anti-viral agents are preferably, e.g., nucleoside analogs (e.g., zidovudine, acyclovir, gancyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin), foscarnet, amantadine, peramivir, rimantadine, saquinavir, indinavir, ritonavir, alpha-interferons and other interferons, AZT, t-705, zanamivir (Relenza ® ), and oseltamivir (Tamiflu ® ).
  • nucleoside analogs e.g., zidovudine, acyclovir, gancyclovir, vidarabine, idoxuridine, trifluridine, and ribavirin
  • foscarnet e.g., amantadine, peramivir, rimantadine, saquinavir,
  • influenza virus vaccines e.g., Fluarix ® (Glaxo SmithKline), FluMist ® (Medlmmune Vaccines), Fluvirin ® (Chiron Corporation), Flulaval ® (GlaxoSmithKline), Afluria ® (CSL Biotherapies Inc.), Agriflu ® (Novartis) or Fluzone ® (Aventis Pasteur).
  • influenza virus vaccines e.g., Fluarix ® (Glaxo SmithKline), FluMist ® (Medlmmune Vaccines), Fluvirin ® (Chiron Corporation), Flulaval ® (GlaxoSmithKline), Afluria ® (CSL Biotherapies Inc.), Agriflu ® (Novartis) or Fluzone ® (Aventis Pasteur).
  • the inventive combination vaccine typically comprises a "safe and effective amount" of the components of the inventive combination vaccine as defined herein.
  • a "safe and effective amount” preferably means an amount of the components, preferably of the at least one RNA encoding at least one F protein or a part thereof of the virus family Paramyxoviridae, and at least one HA protein or a part thereof of the virus family Orthomyxoviridae, that is sufficient to significantly induce a positive modification or prevention of a disease or disorder as defined herein.
  • a "safe and effective amount” is small enough to avoid serious side-effects and to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
  • the invention provides a combination vaccine for use in a method of prophylactic and/or therapeutic treatment of infections caused by viruses of the virus family Paramyxoviridae and/or of the virus family Orthomyxoviridae, particularly respiratory tract infections; preferably RSV infection, mumps, measles, bronchitis, pneumonia, croup, distemper or rinderpest, and influenza. Consequently, in a further aspect, the present invention is directed to the first medical use of the inventive combination vaccine as defined herein as a medicament. Particularly, the invention provides the use of a combination vaccine as defined above for the preparation of a medicament.
  • the present invention is directed to the second medical use of the combination vaccine, as defined herein, optionally in form of a kit or kit of parts, for the treatment of diseases as defined herein.
  • the combination vaccine to be used in a method as said above is a combination vaccine formulated together with a pharmaceutically acceptable vehicle and an optionally additional adjuvant and an optionally additional further component as defined above e.g. a further antigen.
  • the combination vaccine may alternatively be provided such that it is administered for treating diseases as disclosed herein by two doses, each dose containing distinct RNA species, e.g.
  • both doses are administered in a staggered way, i.e. subsequently, shortly one after the other, e.g. within less than 10 minutes, preferably less than 2 minutes, and at the same site of the body to achieve the same immunological effect as for administration of one single composition containing both, e.g. the RNA encoding the the HA protein and the RNA encoding the F protein..
  • the method comprises the in vitro transfection of isolated cells.
  • the cells used therefore are preferably human or animal cells, particularly cells of a primary cell culture, which are then retransferred to a human or animal. Prior to transfection, these cells are typically isolated from the patient to be treated and cultivated.
  • the combination vaccine comprises one or more RNAs encoding for the Fusion (F) protein or a fragment, variant or derivative of the Fusion (F) protein derived from the virus family Paramyxoviridae and for the Hemagglutinin (HA) protein or a fragment, variant or derivative of the Hemagglutinin (HA) protein derived from the virus family Orthomyxoviridae ⁇ o be administered to an individual.
  • both antigenic functions of the combination vaccine are provided by one composition and can be administered to an individual e.g. by one single injection, preferably by parental administration.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, and sublingual injection or infusion techniques.
  • the combination vaccine may be administered to the patient as a single dose.
  • the inventive combination vaccine may be administered to a patient as a single dose followed by a second dose later and optionally even a third, fourth (or more) dose subsequent thereto etc.
  • booster inoculations with the inventive combination vaccine may be administered to a patient at specific time intervals, preferably as defined below, following the second (or third, fourth, etc.) inoculation.
  • such booster inoculations with the inventive combination vaccine may utilize an additional compound or component as defined for the inventive combination vaccine as defined herein.
  • the inventive combination vaccine may be used for human and also for veterinary medical purposes, preferably for human medical purposes. More preferably, the inventive combination vaccine may be used for treating a mammal for diseases as mentioned herein.
  • a mammal may be selected from any mammal, preferably from a mammal, selected from the group comprising, e.g., goat, cattle, swine, dog, cat, donkey, monkey, ape, a rodent such as a mouse, hamster, rabbit, and, in particular, human.
  • Administration modes may be as defined herein.
  • the method comprises the in vitro transfection of isolated cells.
  • a method of treating an individual with his own cells can be performed advantageously by transfection of isolated cells with the inventive combination vaccine.
  • the cells used therefore are preferably human or animal cells, particularly cells of a primary cell culture, which are then retransferred to a human or animal. Prior to transfection, these cells are typically isolated from the patient to be treated and cultivated.
  • the inventive combination vaccine may be administered to the primary cel l culture without further adjuvants or vehicles contained optionally in a combination vaccine according to the invention.
  • the inventive combination vaccine is to be administered to an individual in a pharmaceutically effective amount.
  • the invention is directed to a kit comprising the components of the combination vaccine according to the invention and optionally technical instructions with information on the administration and dosage of the components of the combination vaccine, wherein the combination vaccine contains one or more RNAs (for example mRNAs), said RNAs encoding a first and for a second antigen, wherein the first antigen is a Fusion (F) protein or a fragment, variant or derivative of a Fusion (F) protein derived from the virus family Paramyxoviridae and wherein the second antigen is a Hemagglutinin (HA) protein or a fragment, variant or derivative of a Hemagglutinin (HA) protein derived from the virus family Orthomyxoviridae.
  • RNAs for example mRNAs
  • the first antigen is a Fusion (F) protein or a fragment, variant or derivative of a Fusion (F) protein derived from the virus family Paramyxoviridae
  • the second antigen is a Hemagglutinin (HA) protein or a fragment,
  • the kit may additionally contain a pharmaceutically acceptable vehicle, an adjuvant and at least one further component as defined herein, as well as means for administration and technical instructions.
  • the components of the combination vaccine and e.g. the adjuvant may be provided in lyophilized form.
  • the provided vehicle prior to use of the kit for vaccination, is than added to the lyophilized components in a predetermined amount as written e.g. in the provided technical instructions.
  • the combination vaccine is for use in a method of prophylactic and/or therapeutic treatment of infections caused by viruses of the virus family Paramyxoviridae and/or of the virus family Orthomyxoviridae, particularly respiratory tract infections, e.g. RSV infection, mumps, measles, bronchitis, pneumonia, croup, distemper or rinderpest, and influenza.
  • the invention relates to a combination vaccine as defined herein for use in a method of prophylactic and/or therapeutic treatment of infections caused by viruses of the virus family Paramyxoviridae and/or of the virus family Orthomyxoviridae, particularly respiratory tract infections, e.g.
  • the target group for such treatment preferably comprises infants, particularly pre-term infants (e.g. pre-term neonates), children, the elderly (e.g. people more than 60 years of age, preferably more than 65 years of age) and immunocompromised patients.
  • pre-term infants e.g. pre-term neonates
  • children e.g. people more than 60 years of age, preferably more than 65 years of age
  • immunocompromised patients e.g.
  • the combination vaccine provokes a first adaptive immune response directed against an antigen that is similar or identical to the epitope of a Fusion (F) protein of Paramyxoviridae, and a second immune response elicited by an antigen that is similar or identical to the epitope of a Hemagglutinin (HA) protein of Orthomyxoviridae.
  • F Fusion
  • HA Hemagglutinin
  • composition comprising:
  • RNA preferably mRNA, comprising or consisting of a nucleic acid sequence encoding a protein or peptide, said protein or peptide comprising or consisting of:
  • RNA preferably mRNA, comprising or consisting of a nucleic acid sequence encoding a protein or peptide, said protein or peptide comprising or consisting of: i) the amino acid sequence of a Hemagglutinin (HA) protein of the virus family Orthomyxo viridae,
  • iii) an amino acid sequence exhibiting a sequence identity of at least 80% to said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae of b) i) and/ or said fragment of b) ii).
  • HA Hemagglutinin
  • the Fusion (F) protein of the virus family Paramyxoviridae is a Fusion (F) protein of a virus selected from the group of: Avulavirus, Ferlavirus, Henipavirus, Morbillivirus, Respirovirus, Rubulavirus, TPMV- like viruses, Pneumovirus, Metapneumovirus, Atlantic salmon paramyxovirus, Beilong virus, J virus, Mossman virus, Nariva virus, Salem virus, and Pacific salmon paramyxovirus.
  • the composition according to item 2 wherein the Fusion (F) protein of the virus family Paramyxoviridae s a Fusion (F) protein of a Pneumovirus.
  • composition according to item 3 wherein the Pneumovirus is human respiratory syncytial virus (RSV), preferably RSV Long or RSV A2.
  • RSV human respiratory syncytial virus
  • composition according to item 4 wherein the Fusion (F) protein of the virus family Paramyxoviridae comprises the sequence of SEQ ID No. 1 or SEQ ID No. 2 (or P102A, I379V, and M447V mutants of SEQ ID No. 2).
  • the Hemagglutinin (HA) protein of the virus family Orthomyxoviridae is a Hemagglutinin (HA) protein of an Influenza virus, preferably selected from the group consisting of: Influenza A (e.g. H1 N1 , H1 N2, H2N2, H3N1 , H3N2, H3N8, H5N1 , H5N2, H5N3, H5N8, H5N9, H7N1 , H7N2, H7N3, H7N4, H7N7, H9N1 , H9N2, H10N7), Influenza B, Influenza C, Isavirus (e.g. Infectious salmon anemia virus), Thogotovirus (e.g. Dhori virus), Quaranfil virus, Johnston Atoll virus, and Lake Chad virus.
  • Influenza A e.g. H1 N1 , H1 N2, H2N2, H3N1 , H3N2, H3N8, H5N1 , H5N2,
  • composition according to any of the preceding items, wherein the Hemagglutinin (HA) protein of the virus family Orthomyxoviridae comprises the sequence of SEQ ID No. 3.
  • composition according to any of the preceding items, wherein the composition comprises a combination selected from the group consisting of: RNA of a) i) and the RNA of b) i), RNA of a) i) and the RNA of b) ii), RNA of a) i) and the RNA of b) iii), RNA of a) ii) and the RNA of b) i), RNA of a) ii) and the RNA of b) ii), RNA of a) ii) and the RNA of b) iii), RNA of a) iii) and the RNA of b) i), RNA of a) iii) and the RNA of b) i), RNA of a) iii) and the RNA of b) i), RNA of a) iii) and the RNA of b) i), RNA of a) iii)
  • composition according to item 8 wherein the RNA of a) and the RNA of b) are not the same nucleic acid molecule.
  • composition according to item 10 wherein said nucleic acid molecule does not encode a fusion protein representing a combination of the amino acid sequence of a Fusion (F) protein of the virus family Paramyxoviridae, or fragment thereof with an HA tag of the sequence YPYDVPDYA (SEQ ID No. 22).
  • composition according to any of the preceding items, wherein the composition comprises two or more different RNAs according to a), preferably encoding different peptides comprising the amino acid sequence of different fragments of said Fusion (F) protein of the virus family Paramyxoviridae, wherein most preferably the sequence of all encoded fragments aligned with each other covers the full length of said Fusion (F) protein of the virus family Paramyxoviridae.
  • composition according to any of the preceding items, wherein the composition comprises two or more different RNAs according to b), preferably encoding different peptides comprising the amino acid sequence of different fragments of said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae,, wherein most preferably the sequence of all encoded fragments aligned with each other covers up to the full length of said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae.
  • composition according to any of the preceding items, wherein said fragment of said Fusion (F) protein of the virus family Paramyxoviridae, and/or said fragment of said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae, selected independently of each other, has a length of at least 6 amino acids, preferably at least 7 amino acids, more preferably at least 8 amino acids, even more preferably at least 9 amino acids; even more preferably at least 10 amino acids; even more preferably at least 1 1 amino acids; even more preferably at least 1 2 amino acids; even more preferably at least 1 3 amino acids; even more preferably at least 14 amino acids; even more preferably at least 1 5 amino acids; even more preferably at least 1 6 amino acids; even more preferably at least 1 7 amino acids; even more preferably at least 1 8 amino acids; even more preferably at least 1 9 amino acids; even more preferably at least 20 amino acids; even more preferably at least 25 amino acids; even more preferably at least 30 amino acids; even more preferably at least 35 amino acids;
  • composition according to any of the preceding items, wherein said fragment of said Fusion (F) protein of the virus family Paramyxoviridae, and/or said fragment of said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae, is selected independently of each other from the first, second, third or fourth quarter of the amino acid sequence of said Fusion (F) protein of the virus family Paramyxoviridae and/or the amino acid sequence of said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae, respectively.
  • composition according to any of the preceding items, wherein the sequence identity of a) iii) and/or b) iii), selected independently of each other, is at least 85%, preferably at least 90%, even more preferably at least 92%; even more preferably at least 92%; even more preferably at least 94%; even more preferably at least 95%; even more preferably at least 96%; even more preferably at least 97%; even more preferably at least 98%; most preferably at least 99%.
  • composition according to any of the preceding items, with the proviso that if the RNA of a) and the RNA of b) are the same nucleic acid molecule and the protein or peptide of a) and peptide of b) encoded by said same nucleic acid are the same protein or peptide, and said same protein or peptide comprises the sequence of SEQ ID No. 22, then said same nucleic acid molecule encodes a protein or peptide comprising:
  • an amino acid sequence of b) ii), said fragment having a length of at least 10 amino acids, preferably at least 1 1 amino acids, more preferably at least 12 amino acids, more preferably at least 13 amino acids, more preferably at least 14 amino acids, more preferably at least 15 amino acids, more preferably at least 1 6 amino acids, more preferably at least 1 7 amino acids, more preferably at least 18 amino acids, more preferably at least 19 amino acids, most preferably at least 20 amino acids;
  • an amino acid sequence exhibiting a sequence identity of at least 80% to of a fragment of an Hemagglutinin (HA) protein of the virus family Orthomyxoviridae, said fragment having a length of at least 12 amino acids; and/or
  • the composition comprises at least two monocistronic RNAs, wherein the composition is selected from the group: (a) at least one monocistronic RNA according to SEQ ID No.: 13 and at least one monocistronic RNA according to SEQ ID No.: 18 or SEQ ID No.: 21 , (b) at least one monocistronic RNA according to SEQ ID No.: 14 and at least one monocistronic RNA according to SEQ ID No.: 18 or according to SEQ ID No.: 21 , (c) at least one monocistronic RNA according to SEQ ID No.: 15 and at least one monocistronic RNA according to SEQ ID No.: 18 or SEQ ID No.: 21 , (d) at least one monocistronic RNA according to SEQ ID No.: 16 and at least one monocistronic RNA according to SEQ ID No.: 18 or SEQ ID No.: 21 , (e) at least one monocistronic RNA according to
  • composition according to any of the preceding items wherein the RNA of b) comprises or consists of a nucleic acid sequence selected from the group consisting of SEQ ID No: 1 8 and SEQ ID No. 21 .
  • the RNA of a) comprises or consists of the sequence of SEQ ID No. 1 9 and/or SEQ ID No. 20;
  • the RNA of b) comprises or consists of the sequence of SEQ ID No. 21 .
  • a histone-stem-loop structure preferably a histone-stem-loop in its 3' untranslated region
  • composition according to any of the preceding items wherein the RNA of a) and/or the RNA of b) are associated with or complexed with a cationic or polycationic compound or a polymeric carrier, optionally in a weight ratio selected independently of each other from a range of about 6:1 (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/w) to about 0.5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w:w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ration of about 3:1 (w/w) to about 2:1 (w/w) of nucleic to cationic or polycationic compound and/or with a polymeric carrier; or optionally in a nitrogen/phosphate ratio of nucleic to cationic or polycationic compound and/or polymeric carrier in the range of about 0.1 -10, preferably in a range of about a
  • composition according to any of the preceding items, wherein the RNA of a) and/or the RNA of b) are associated or complexed with a cationic protein or peptide, preferably protamine.
  • composition further comprises an adjuvant; preferably an adjuvant comprising or consisting of an oligo- or a polynucleotide; more preferably an adjuvant comprising or consisting of a RNA or a DNA; even more preferably an adjuvant comprising or consisting of a RNA or a DNA, said RNA or DNA being complexed with a cationic or polycationic compound and/or with a polymeric carrier; optionally in a weight ratio selected from a range of about 6:1 (w/w) to about 0.25:1 (w/w), more preferably from about 5:1 (w/w) to about 0.5:1 (w/w), even more preferably of about 4:1 (w/w) to about 1 :1 (w:w) or of about 3:1 (w/w) to about 1 :1 (w/w), and most preferably a ration of about 3:1 (w/w) to about 2:1 (w/w) of adjuvant component to
  • composition according to any of the preceding items, wherein the composition further comprises an auxiliary substance selected from lipopolysaccharides, TNF- alpha, CD40 ligand, or cytokines, monokines, lymphokines, interleukins or chemokines, IL-1 , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-14, IL-1 5, IL-16, IL-1 7, IL-18, IL-19, IL-20, IL-21 , IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31 , IL-32, IL-33, IFN-alpha, IFN-beta, IFN-gamma, GM-CSF, G-CSF, M-CSF, LT-beta, TNF-alpha
  • composition according to any of the preceding items wherein the composition is a pharmaceutical composition optionally further comprising a pharmaceutically acceptable carrier, excipient or diluent.
  • composition according to item 33 wherein the composition is formulated for parenteral, oral, nasal, pulmonary, topical, rectal, buccal, or vaginal administration or for administration by inhalation or via an implanted reservoir.
  • composition according to any of items 1 to 34 for use in a method of
  • the composition according to any of items 35 to 37, wherein the method comprises the in v/ ' t o transfection of isolated cells.
  • the composition according to any of items 35 to 38, wherein the individual to be treated is selected from the group consisting of infants, particularly pre-term neonates, children, the elderly and immunocompromised patients. Kit comprising:
  • RNA preferably mRNA, comprising or consisting of a nucleic acid sequence encoding a protein or peptide, said protein or peptide comprising or consisting of:
  • RNA preferably mRNA, comprising or consisting of a nucleic acid sequence encoding a protein or peptide, said protein or peptide comprising or consisting of:
  • HA Hemagglutinin
  • RNA of a) and the RNA of b) are as defined in any of items 2 to 34.
  • the kit according to item 40 or 41 for use in a method of prophylactic and/or therapeutic treatment of the human or animal body.
  • the kit according to any of items 40 to 44, wherein the method comprises the in vitto transfection of isolated cells.
  • the kit according to any of items 40 to 45, wherein the individual to be treated is selected from the group consisting of infants, particularly pre-term neonates, children, the elderly and immunocompromised patients.
  • the method to item 47, wherein said composition or kit is used as vaccine.
  • the method according to item 47 or 48, wherein the method comprises the in vitro transfection of isolated cells.
  • nucleic acid comprising or consisting of a sequence selected from the group of: SEQ ID No. 7, SEQ ID No. 8, SEQ ID No. 9, SEQ ID No. 10, SEQ ID No. 1 1 , and SEQ ID No: 12.
  • Nucleic acid comprising or consisting of a sequence selected from the group of: SEQ ID No. 13, SEQ ID No. 14, SEQ ID No. 15, SEQ ID No. 16, SEQ ID No. 1 7, and SEQ ID No: 18.
  • Nucleic acid according to item 52 the nucleic acid comprising or consisting of a sequence selected from the group of: SEQ ID No. 19, SEQ ID No. 20 and SEQ ID No. 21 .
  • RNA preferably mRNA, comprising or consisting of a nucleic acid sequence encoding a protein or peptide, said protein or peptide comprising or consisting of: i) the amino acid sequence of a Fusion (F) protein of the virus family Paramyxoviridae;
  • RNA of item 54 wherein the RNA is as defined in any of items 2 to 5, 9 to 13, 16 to 21 , or 25 to 32, in particular wherein the RNA comprises or consists of a sequence selected from the group consisting of SEQ ID No. 1 3, SEQ ID No. 14, SEQ ID No. 15, SEQ ID No. 1 6, SEQ ID No. 1 7, SEQ ID No. 1 9 and SEQ ID No. 20.
  • RNA preferably mRNA, comprising or consisting of a nucleic acid sequence encoding a protein or peptide, said protein or peptide comprising or consisting of: i) the amino acid sequence of a Hemagglutinin (HA) protein of the virus family Orthomyxo viridae,
  • HA Hemagglutinin
  • iii) an amino acid sequence exhibiting a sequence identity of at least 80% to said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae o b) i) and/ or said fragment of b) ii); for use in a method according to any of items 47 to 50.
  • HA Hemagglutinin
  • RNA of item 56 wherein the RNA is as defined in any of items 6 to 7, 9 to 13, 1 6 to 20, 22, or 25 to 32, in particular wherein the RNA comprises or consists of a sequence selected from the group consisting of SEQ ID No: 18 or SEQ ID No. 21 .
  • an HA-tag is preferably not used according to the invention as fragment of said Hemagglutinin (HA) protein of the virus family Orthomyxoviridae.
  • RNA encoding a mere HA-tagged Fusion (F) proteins of the virus family Paramyxoviridae, e.g. a Fusion (F) protein of the virus family Paramyxoviridae linked (optionally via a short 1 to 10 amino acid peptide linker) to an HA tag - or nucleic acids encoding the same - is not an preferred embodiment of composition of the invention.
  • F Fusion
  • the encoded fusion protein must preferably comprise as Hemagglutinin (HA) derived portion (peptide b in item 1 ) aside of the HA tag preferably also other sequence elements of an Hemagglutinin protein.
  • the Hemagglutinin (HA) derived portion may be longer 63
  • HA Hemagglutinin
  • An additional HA tag i.e. in addition to another non-HA sequence qualifying as Hemagglutinin (HA) derived portion (see peptide b in item 1 ), is also possible.
  • the fusion of fragments of an Fusion (F) protein - instead of the full length F>Uion (F) protein - with an HA tag is not excluded from the scope of the present invention.
  • a fusion protein of an Fusion (F) protein as defined herein with an HA tag is furthermore particularly acceptable, when the composition comprises - aside of said fusion protein - another distinct peptide which fulfils the requirements of peptide b in item 1 in lieu thereof.
  • RNA encoding an HA tagged Fusion (F) protein is clearly contemplated by the present invention, for example as vaccine or in a method of treatment as disclosed herein.
  • Figure 1 shows the induction of a RSV F protein specific CTL immune response in
  • mice after vaccination with mRNA coding for RSV Long F protein and mRNA coding for HA (Hemagglutinin of A/Puerto Rico/8/34), injected in one pharmaceutical composition (F (RSV Long) + HA cocktail) or separately injected (F (RSV Long) + HA sep. injected).
  • F (RSV Long) + HA cocktail mRNA coding for F protein
  • F (RSV Long) + HA sep. injected To control for unspecific immune effects of the cocktail application, one group was treated with a cocktail of mRNA coding for F protein (RSV Long) and a non-coding RNA.
  • mice were treated with buffer.
  • One week after the last vaccination antigen specific T cells were analysed in spleens of vaccinated mice by ELISPOT analysis.
  • Splenocytes were either stimulated with an ⁇ -2 ⁇ - restricted T-cell epitope of the F protein KYKNAVTEL (amino acids 85-93; SEQ ID No. 24) or DMSO alone. Lines represent the median. Statistical analysis was done by the Mann-Whitney test.
  • Figure 2 shows the induction of a RSV F protein specific CTL immune response in
  • mice BALB/c mice after vaccination with mRNA coding for RSV A2 F protein and mRNA coding for HA (Hemagglutinin of A/Puerto Rico/8/34), injected in one pharmaceutical composition (F (RSV A2) + HA cocktail) or separately injected (F (RSV A2) + HA sep. injected).
  • F (RSV A2) + HA cocktail) or separately injected
  • F (RSV A2) + HA sep. injected mice were treated with buffer.
  • antigen specific T cells were analysed in spleens of vaccinated mice by ELISPOT analysis. Splenocytes were either stimulated with three H-2k d -restricted T-cell epitopes of the F protein (KYKNAVTEL (amino acids 85-93; SEQ ID No.
  • the combination of both coding mRNAs in one pharmaceutical composition increases the induction of F protein specific antibodies in 3 of 5 mice compared to the group vaccinated with the combination of mRNA coding for F protein and non-coding RNA.
  • Mice either received the two mRNAs at separate injection sites (F+HA sep. injected) or as a cocktai l of both mRNAs. For negative control, mice were treated with buffer. Four weeks after second immunization antigen specific antibodies were analysed in serum of vaccinated mice. Lines represent the median.
  • FIG. 7 shows the wild type coding sequence of Fusion (F) protein of RSV Long (Human respiratory syncytial virus strain ATCC VR-26 (NCBI Accession No. AY91 1262) according to SEQ ID No. 4. shows the wild type coding sequence of Fusion (F) protein of RSV A2 (NCBI Accession No. M1 1486.1 ) according to SEQ ID No. 5. shows the wild type coding sequence of Hemagglutinin (HA) protein of Influenza A/Puerto Rico/8/1 934 (NCBI Accession No. EF467821 ) according to SEQ ID No. 6. shows the GC-enriched DNA coding sequence (SEQ ID No. 7) coding for the Fusion (F) protein of RSV Long.
  • FIG. 8 shows the GC-enriched DNA coding sequence (SEQ ID No. 8) coding for the Fusion (F) protein of RSV A2.
  • FIG. 12 shows the GC-enriched DNA coding sequence (SEQ ID No: 12) coding for the Hemagglutinin (HA) protein of Influenza A/Puerto Rico/8/1934. shows an GC-enriched RNA sequence without UTRs (SEQ ID No. 13) coding for the Fusion (F) protein of RSV Long. shows an GC-enriched RNA sequence without UTRs (SEQ ID No. 14) coding for the Fusion (F) protein of RSV A2. shows an GC-enriched RNA sequence without UTRs (SEQ ID No. 1 5) coding for the Fusion (F) protein of RSV A2 (P102A). shows an GC-enriched RNA sequence without UTRs (SEQ ID No.
  • FIG. 21 shows the GC-enriched full mRNA sequence coding for the Hemagglutinin (HA) protein of Influenza A Puerto Rico/8/1934 according to SEQ ID No. 21 . shows the non-coding RNA according to SEQ ID No. 23 used as a control.
  • DNA sequences encoding the F protein of RSV-Long (SEQ ID No. 1 ), RSV-A2 (SEQ ID No. 2) and Hemagglutinin of A/Puerto Rico/8/34 (HA) (SEQ ID No. 3), and non-coding RNA as control (SEQ ID No. 23), were prepared and used for subsequent in vitro transcription reactions.
  • All used DNA sequences (SEQ ID No. 7, SEQ ID No. 8 and SEQ ID No: 12) were prepared by modifying the wild type encoding DNA sequences by introducing a GC-optimized sequence for a better codon usage and stabilization.
  • SEQ ID No. 19 SEQ ID No. 20 and SEQ ID No. 21 the sequences of the corresponding mRNAs are shown.
  • sequences was furthermore introduced into a pCV19 vector and modified to comprise stabilizing sequences derived from alpha-globin-3'-UTR (muag (mutated alpha-globin-3'-UTR)), a histone-stem- loop structure, and a stretch of 70 x adenosine at the 3'-terminal end (poly-A-tail).
  • the respective DNA plasmids prepared above were transcribed into mRNA in vitro using T7-Polymerase. Subsequently the obtained mRNA was purified using PureMessenger ® (CureVac, Tubingen, Germany).
  • mRNA complexation consisted of a mixture of 50% free mRNA and 50% mRNA complexed with protamine at a weight ratio of 2:1 .
  • mRNA was complexed with protamine by slow addition of protamine- Ringer's lactate solution to mRNA. As soon as the complexes were stably generated, free mRNA was added, stirred shortly and the final concentration of the vaccine was adjusted with Ringer's lactate solution.
  • mice were vaccinated twice intradermally with the vaccine comprising 80 pg mRNA coding for HA (Hemagglutinin of A/Puerto Rico/8/34) and 80 pg mRNA coding for F protein (RSV-A2).
  • Mice either received the two mRNAs at separate injection sites (F (RSV A2) + HA sep. injected) or as a cocktail of both mRNAs (F (RSV A2) + HA cocktail).
  • F (RSV A2) + HA sep. injected or as a cocktail of both mRNAs
  • mice were treated with buffer.
  • mice were vaccinated twice intradermally with the vaccine comprising 10 ⁇ g mRNA coding for HA (Hemagglutinin of A/Puerto Rico/8/34) and 10 pg mRNA coding for F protein (RSV-Long).
  • Mice either received the two mRNAs at separate injection sites (F (RSV Long) + HA sep. injected) or as a cocktail of both mRNAs (F (RSV Long) + HA cocktail.
  • F (RSV Long) + HA cocktail To control for unspecific immune effects of the cocktail application, one group was treated with a cocktail of F (RSV Long) mRNA and a non-coding RNA (F (RSV Long) + non-coding RNA).
  • mice were treated with buffer.
  • Detection of an antigen specific immune response was carried out by detecting RSV F protein or HA protein specific antibodies. Therefore, blood samples were taken from vaccinated mice two and four weeks after the last vaccination and sera were prepared. MaxiSorp ® plates (Nalgene Nunc International) were coated with F (Sino Biological Inc.) or HA protein (Charles River Laboratories). After blocking with I xPBS containing 0.05% Tween-20 and 1 % BSA the plates were incubated with diluted mouse serum (1 :50). Subsequently a biotin-coupled secondary antibody (Anti-mouse-lgG Dianova, cat. #1 15035003) was added.
  • mice immunized with HA and RSV-A2, example 2 Four weeks (mice immunized with HA and RSV-A2, example 2) or one week (mice immunized with HA and RSV-Long, example 3) after the last vaccination mice were sacrificed, the spleens were removed and the splenocytes were isolated.
  • a coat multiscreen plate (Millipore) was incubated overnight with coating buffer 0.1 M Carbonat-Bicarbonat Buffer pH 9.6, 10.59 g/l Na 2 C0 3 , 8.4g/l NaHCOj) comprising antibody against INFy (BD Pharmingen, Heidelberg, Germany).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Pulmonology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

La présente invention concerne un vaccin, en particulier un vaccin combiné présentant au moins une première et une deuxième fonction antigénique, le vaccin combiné comprenant au moins un ARN codant pour au moins un(e) ou plusieurs protéines ou fragments, variants ou dérivés de protéines décernant une fonction antigénique, la première fonction antigénique étant une protéine de fusion (F) ou un fragment, un variant ou un dérivé d'une protéine de fusion (F) dérivée de la famille des virus Paramyxoviridae et la deuxième fonction antigénique étant une protéine d'hémagglutinine (HA) ou un fragment, un variant ou un dérivé d'une protéine d'hémagglutinine (HA) dérivée de la famille des virus Orthomyxoviridae. De plus, la présente invention concerne un kit ou un kit de parties comprenant les constituants dudit vaccin combiné et ledit vaccin combiné destiné à être utilisé dans un procédé de traitement prophylactique ou thérapeutique de maladies, en particulier dans la prévention ou le traitement de maladies infectieuses telles que le VRS et la grippe.
PCT/EP2014/002302 2013-08-21 2014-08-21 Vaccin combiné WO2015024669A1 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CA2915730A CA2915730A1 (fr) 2013-08-21 2014-08-21 Vaccin combine contre le virus respiratoire syncytial (rsv) et la grippea
AU2014310935A AU2014310935B2 (en) 2013-08-21 2014-08-21 Combination vaccine
EP14761282.4A EP3035959A1 (fr) 2013-08-21 2014-08-21 Vaccin combiné
CN201480044258.XA CN105473157A (zh) 2013-08-21 2014-08-21 组合疫苗
US15/048,561 US10588959B2 (en) 2013-08-21 2016-02-19 Combination vaccine
US16/781,781 US11266735B2 (en) 2013-08-21 2020-02-04 Combination vaccine
US17/590,173 US20220152193A1 (en) 2013-08-21 2022-02-01 Combination vaccine

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP2013002513 2013-08-21
EPPCT/EP2013/002513 2013-08-21

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/048,561 Continuation US10588959B2 (en) 2013-08-21 2016-02-19 Combination vaccine

Publications (1)

Publication Number Publication Date
WO2015024669A1 true WO2015024669A1 (fr) 2015-02-26

Family

ID=51492914

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/002302 WO2015024669A1 (fr) 2013-08-21 2014-08-21 Vaccin combiné

Country Status (5)

Country Link
US (3) US10588959B2 (fr)
CN (1) CN105473157A (fr)
AU (1) AU2014310935B2 (fr)
CA (1) CA2915730A1 (fr)
WO (1) WO2015024669A1 (fr)

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016011401A1 (fr) * 2014-07-18 2016-01-21 Kedl Ross M Combinaisons d'immunostimulation et leur utilisation
WO2017049245A2 (fr) 2015-09-17 2017-03-23 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2017066793A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffes arnm et procédés de coiffage d'arnm
WO2017066791A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm à substitution sucre
WO2017066781A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm à liaison phosphate modifié
WO2017066789A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm avec sucre modifié
WO2017066782A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffes d'arnm hydrophobes
WO2017112865A1 (fr) 2015-12-22 2017-06-29 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques et/ou prophylactiques
WO2017109134A1 (fr) * 2015-12-22 2017-06-29 Curevac Ag Procédé de production de compositions de molécules d'arn
WO2017070620A3 (fr) * 2015-10-22 2017-07-13 Modernatx, Inc. Vaccin contre le virus de la grippe à large spectre
WO2017191258A1 (fr) * 2016-05-04 2017-11-09 Curevac Ag Vaccins contre l'arnm de la grippe
WO2017218704A1 (fr) 2016-06-14 2017-12-21 Modernatx, Inc. Formulations stabilisées de nanoparticules lipidiques
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
WO2018089540A1 (fr) 2016-11-08 2018-05-17 Modernatx, Inc. Formulations stabilisées de nanoparticules lipidiques
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
WO2018115507A3 (fr) * 2016-12-23 2018-08-30 Curevac Ag Vaccin contre l'hénipavirus
WO2018170336A1 (fr) 2017-03-15 2018-09-20 Modernatx, Inc. Formulation de nanoparticules lipidiques
WO2018170306A1 (fr) 2017-03-15 2018-09-20 Modernatx, Inc. Composés et compositions d'administration intracellulaire d'agents thérapeutiques
WO2018232120A1 (fr) 2017-06-14 2018-12-20 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents
WO2019036638A1 (fr) 2017-08-18 2019-02-21 Modernatx, Inc. Procédés de préparation d'arn modifié
WO2019046809A1 (fr) 2017-08-31 2019-03-07 Modernatx, Inc. Procédés de fabrication de nanoparticules lipidiques
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2019148101A1 (fr) * 2018-01-29 2019-08-01 Modernatx, Inc. Vaccins à base d'arn contre le vrs
WO2020061367A1 (fr) 2018-09-19 2020-03-26 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2020061457A1 (fr) 2018-09-20 2020-03-26 Modernatx, Inc. Préparation de nanoparticules lipidiques et leurs méthodes d'administration
WO2020160397A1 (fr) 2019-01-31 2020-08-06 Modernatx, Inc. Procédés de préparation de nanoparticules lipidiques
WO2020160430A1 (fr) 2019-01-31 2020-08-06 Modernatx, Inc. Mélangeurs à tourbillon et procédés, systèmes, et appareils associés
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
EP3595676A4 (fr) * 2017-03-17 2021-05-05 Modernatx, Inc. Vaccins à base d'arn contre des maladies zoonotiques
WO2021204179A1 (fr) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Vaccins à base d'acide nucléique pour coronavirus
WO2021204175A1 (fr) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Compositions de nanoparticules lipidiques
WO2022002040A1 (fr) 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2022037652A1 (fr) 2020-08-20 2022-02-24 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2022152141A2 (fr) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques conjugués polymères et compositions de nanoparticules lipidiques
WO2022152109A2 (fr) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
EP4035659A1 (fr) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes destinés à l'administration d'agents thérapeutiques
US11433139B2 (en) 2018-03-16 2022-09-06 Zoetis Services Llc Peptide vaccines against interleukin-31
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
WO2022247755A1 (fr) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
WO2023044333A1 (fr) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Lipides cycliques et leurs procédés d'utilisation
WO2023044343A1 (fr) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Lipides acycliques et leurs procédés d'utilisation
EP4162950A1 (fr) 2021-10-08 2023-04-12 Suzhou Abogen Biosciences Co., Ltd. Vaccins d'acide nucléique pour coronavirus
WO2023056917A1 (fr) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2023056914A1 (fr) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US11667910B2 (en) 2015-05-29 2023-06-06 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
WO2023122752A1 (fr) 2021-12-23 2023-06-29 Renagade Therapeutics Management Inc. Lipides contraints et procédés d'utilisation associés
US11739125B2 (en) 2013-08-21 2023-08-29 Cure Vac SE Respiratory syncytial virus (RSV) vaccine
WO2023196931A1 (fr) 2022-04-07 2023-10-12 Renagade Therapeutics Management Inc. Lipides cycliques et nanoparticules lipidiques (npl) pour l'apport d'acides nucléiques ou de peptides destinés à être utilisés dans la vaccination contre des agents infectieux
US11786590B2 (en) 2015-11-09 2023-10-17 CureVac SE Rotavirus vaccines
US11865084B2 (en) 2016-12-23 2024-01-09 CureVac SE MERS coronavirus vaccine
WO2024037578A1 (fr) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition de nanoparticules lipidiques
WO2024089634A1 (fr) * 2022-10-27 2024-05-02 Pfizer Inc. Compositions immunogènes contre la grippe et le vrs
US11975064B2 (en) 2011-03-02 2024-05-07 CureVac SE Vaccination with mRNA-coded antigens

Families Citing this family (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013120497A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour l'augmentation de l'expression d'une protéine thérapeutique codée
SG11201506052PA (en) 2013-02-22 2015-09-29 Curevac Gmbh Combination of vaccination and inhibition of the pd-1 pathway
WO2015024669A1 (fr) 2013-08-21 2015-02-26 Curevac Gmbh Vaccin combiné
AU2014310931B2 (en) 2013-08-21 2019-12-19 CureVac SE Rabies vaccine
SG10201801428RA (en) 2013-08-21 2018-03-28 Curevac Ag Method for increasing expression of rna-encoded proteins
ES2806575T3 (es) 2013-11-01 2021-02-18 Curevac Ag ARN modificado con propiedades inmunoestimuladoras disminuidas
WO2015101415A1 (fr) 2013-12-30 2015-07-09 Curevac Gmbh Molécules d'acides nucléiques artificielles
SG10201805660WA (en) 2013-12-30 2018-08-30 Curevac Ag Methods for rna analysis
WO2015149944A2 (fr) 2014-04-01 2015-10-08 Curevac Gmbh Complexe cargo de support polymère à utiliser comme agent immunostimulant ou comme adjuvant
ES2727776T3 (es) 2014-06-10 2019-10-18 Curevac Ag Método para mejorar la producción de ARN
RS63848B1 (sr) 2014-06-25 2023-01-31 Acuitas Therapeutics Inc Novi lipidi i formulacije lipidnih nanočestica za isporuku nukleinskih kiselina
PT3708668T (pt) 2014-12-12 2022-09-14 Curevac Ag Moléculas de ácido nucleico artificiais para uma expressão melhorada de proteína
US10653768B2 (en) 2015-04-13 2020-05-19 Curevac Real Estate Gmbh Method for producing RNA compositions
EP4026568A1 (fr) 2015-04-17 2022-07-13 CureVac Real Estate GmbH Lyophilisation de l'arn
DK3326641T3 (da) 2015-04-22 2019-09-30 Curevac Ag Rna-holdig sammensætning til behandling af tumorsygdomme
US11384375B2 (en) 2015-04-30 2022-07-12 Curevac Ag Immobilized poly(n)polymerase
US11559570B2 (en) 2015-05-15 2023-01-24 CureVac SE Prime-boost regimens involving administration of at least one mRNA construct
EP3297682B1 (fr) 2015-05-20 2021-07-14 CureVac AG Composition de poudre sèche comprenant de l'arn à chaîne longue
US10517827B2 (en) 2015-05-20 2019-12-31 Curevac Ag Dry powder composition comprising long-chain RNA
EP4098743A1 (fr) 2015-05-29 2022-12-07 CureVac AG Procédé pour ajouter des structures de capuchon à l'arn à l'aide d'enzymes immobilisés
CN107922364B (zh) 2015-06-29 2021-12-31 爱康泰生治疗公司 用于递送核酸的脂质和脂质纳米颗粒制剂
US10501768B2 (en) 2015-07-13 2019-12-10 Curevac Ag Method of producing RNA from circular DNA and corresponding template DNA
EP3362576A1 (fr) 2015-10-12 2018-08-22 CureVac AG Procédé automatisé d'isolement, de sélection et/ou de détection de micro-organismes ou de cellules présents dans une solution
MD3718565T2 (ro) 2015-10-22 2022-09-30 Modernatx Inc Vaccinuri împotriva virusului respirator
JP7030690B2 (ja) 2015-10-28 2022-03-07 アキィタス・セラピューティクス・インコーポレイテッド 核酸のデリバリーのための新規脂質および脂質ナノ粒子製剤
US11248223B2 (en) 2015-12-23 2022-02-15 Curevac Ag Method of RNA in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
MX2018009917A (es) 2016-02-17 2019-08-14 Curevac Ag Vacuna contra el virus del zika.
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
US10538786B2 (en) 2016-04-13 2020-01-21 Janssen Pharmaceuticals, Inc. Recombinant arterivirus replicon systems and uses thereof
CN105693829B (zh) * 2016-04-14 2022-03-15 青岛易邦生物工程有限公司 一种b型禽偏肺病毒f蛋白
US11596699B2 (en) 2016-04-29 2023-03-07 CureVac SE RNA encoding an antibody
EP3452493A1 (fr) 2016-05-04 2019-03-13 CureVac AG Molécules d'acide nucléique et leurs utilisations
EP3452101A2 (fr) 2016-05-04 2019-03-13 CureVac AG Arn codant pour une protéine thérapeutique
EP3468613A1 (fr) 2016-06-09 2019-04-17 CureVac AG Supports hybrides pour cargo d'acides nucléiques
CN105969741A (zh) * 2016-06-15 2016-09-28 湖北省农业科学院畜牧兽医研究所 共表达h9亚型禽流感病毒ha和鸡白介素6蛋白的重组新城疫耐热疫苗株及制备方法
WO2018010815A1 (fr) * 2016-07-15 2018-01-18 Biontech Rna Pharmaceuticals Gmbh Formule pour l'administration d'arn.
KR20190082226A (ko) 2016-10-17 2019-07-09 신테틱 제노믹스, 인코포레이티드. 재조합 바이러스 레플리콘 시스템 및 그의 용도
WO2018096179A1 (fr) 2016-11-28 2018-05-31 Curevac Ag Procédé de purification d'arn
BR112019011661A2 (pt) 2016-12-05 2020-01-07 Synthetic Genomics, Inc. Composições e métodos para aumentar expressão de gene
JP2020501545A (ja) 2016-12-08 2020-01-23 キュアバック アーゲー 肝疾患の処置または予防のためのrna
WO2018104540A1 (fr) 2016-12-08 2018-06-14 Curevac Ag Arn pour la cicatrisation des plaies
US11464847B2 (en) 2016-12-23 2022-10-11 Curevac Ag Lassa virus vaccine
CN110914433A (zh) 2017-03-24 2020-03-24 库尔维科公司 编码crispr相关蛋白质的核酸及其用途
US11357856B2 (en) 2017-04-13 2022-06-14 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
CN107961371B (zh) * 2017-04-19 2020-08-11 武汉博沃生物科技有限公司 季节流感-rsv联合疫苗及其制备方法和应用
US11596679B2 (en) 2017-04-27 2023-03-07 Vanderbilt University Hepatitis C virus gene sequences and methods of use therefor
AU2018256877B2 (en) 2017-04-28 2022-06-02 Acuitas Therapeutics, Inc. Novel carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
AU2018293925A1 (en) * 2017-06-30 2020-01-16 Monash University A method of treatment
CN111328287A (zh) 2017-07-04 2020-06-23 库瑞瓦格股份公司 新型核酸分子
WO2019036008A1 (fr) 2017-08-16 2019-02-21 Acuitas Therapeutics, Inc. Lipides destinés à être utilisés dans des formulations nanoparticulaires lipidiques
WO2019036030A1 (fr) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipides destinés à être utilisés dans des formulations de nanoparticules lipidiques
WO2019036028A1 (fr) 2017-08-17 2019-02-21 Acuitas Therapeutics, Inc. Lipides destinés à être utilisés dans des formulations nanoparticulaires lipidiques
WO2019038332A1 (fr) 2017-08-22 2019-02-28 Curevac Ag Vaccin contre les bunyavirus
WO2019092153A1 (fr) 2017-11-08 2019-05-16 Curevac Ag Adaptation de séquence d'arn
US11931406B2 (en) 2017-12-13 2024-03-19 CureVac SE Flavivirus vaccine
EA202091517A1 (ru) 2017-12-19 2020-11-03 Янссен Сайенсиз Айрлэнд Анлимитед Компани Способы и устройство для доставки вакцин против вируса гепатита b (hbv)
US11020476B2 (en) 2017-12-19 2021-06-01 Janssen Sciences Ireland Unlimited Company Methods and compositions for inducing an immune response against Hepatitis B Virus (HBV)
US11021692B2 (en) 2017-12-19 2021-06-01 Janssen Sciences Ireland Unlimited Company Hepatitis B virus (HBV) vaccines and uses thereof
EP3728634A1 (fr) 2017-12-21 2020-10-28 CureVac AG Adn linéaire double brin couplé à un support ou une étiquette unique et procédés de production dudit adn linéaire double brin
WO2019143949A2 (fr) 2018-01-19 2019-07-25 Synthetic Genomics, Inc. Induction et amélioration des réponses immunitaires à l'aide de systèmes de réplicon de recombinaison
CN108300806A (zh) * 2018-01-29 2018-07-20 广西大学 一种快速检测Ferlavirus病毒的RT-PCR引物及其应用
CN109852588B (zh) * 2018-12-24 2023-03-07 中国水产科学研究院珠江水产研究所 一种抗罗非鱼免疫球蛋白IgM的单克隆抗体及其细胞株和应用
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
MX2022012251A (es) * 2020-03-31 2023-03-13 Univ Pennsylvania Vacuna universal contra la influenza usando arnm modificado con nucleósidos.
JP2023527422A (ja) * 2020-05-28 2023-06-28 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン SARS-CoV-2感染を予防および治療するための組成物および方法
EP4182297A1 (fr) 2020-07-16 2023-05-24 Acuitas Therapeutics, Inc. Lipides cationiques destinés à être utilisés dans des nanoparticules lipidiques
CN112402603A (zh) * 2020-12-02 2021-02-26 林建宏 气雾免疫用的温敏型佐剂及其制作方法
CA3205569A1 (fr) 2020-12-22 2022-06-30 CureVac SE Vaccin a arn contre des variants sras-cov-2
WO2022212659A1 (fr) * 2021-03-31 2022-10-06 The Trustees Of The University Of Pennsylvania Compositions de vaccin à arnm multigénique et procédés d'utilisation
CN113082187B (zh) * 2021-04-09 2023-06-27 北京红太阳药业有限公司 小柴胡提取物与阿奇霉素联合在治疗病毒性肺炎中的应用
CN116694568A (zh) * 2022-03-02 2023-09-05 北京市希波生物医学技术有限责任公司 用于激活整体抗肿瘤免疫系统的培养基配方物及制备激动剂激活的整体免疫效应细胞的方法
CN115137814A (zh) * 2022-07-01 2022-10-04 可蓝赛生物医药(上海)有限公司 一种肿瘤疫苗佐剂
CN116650632B (zh) * 2023-03-31 2023-10-24 北京吉诺卫生物科技有限公司 一种流感病毒和rsv的联合疫苗、其制备方法与应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011008974A2 (fr) * 2009-07-15 2011-01-20 Novartis Ag Compositions à base de protéine f du vrs et procédés de fabrication associés
WO2012116811A1 (fr) * 2011-03-02 2012-09-07 Curevac Gmbh Vaccination chez des patients âgés

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2305699B1 (fr) 2001-06-05 2014-08-13 CureVac GmbH ARNm stabilisée avec un contenu augmenté en G/C et optimisée pour la translation dans ses zones codées pour la vaccination contre la trypanosomiase, la leishmaniose et la toxoplasmose
DE10162480A1 (de) 2001-12-19 2003-08-07 Ingmar Hoerr Die Applikation von mRNA für den Einsatz als Therapeutikum gegen Tumorerkrankungen
DE10229872A1 (de) 2002-07-03 2004-01-29 Curevac Gmbh Immunstimulation durch chemisch modifizierte RNA
DE10335833A1 (de) 2003-08-05 2005-03-03 Curevac Gmbh Transfektion von Blutzellen mit mRNA zur Immunstimulation und Gentherapie
DE102004042546A1 (de) 2004-09-02 2006-03-09 Curevac Gmbh Kombinationstherapie zur Immunstimulation
DE102005023170A1 (de) 2005-05-19 2006-11-23 Curevac Gmbh Optimierte Formulierung für mRNA
DE102006035618A1 (de) 2006-07-31 2008-02-07 Curevac Gmbh Nukleinsäure der Formel (I): GlXmGn, insbesondere als immunstimulierendes Adjuvanz
DE102006061015A1 (de) 2006-12-22 2008-06-26 Curevac Gmbh Verfahren zur Reinigung von RNA im präparativen Maßstab mittels HPLC
DE102007001370A1 (de) 2007-01-09 2008-07-10 Curevac Gmbh RNA-kodierte Antikörper
WO2009030254A1 (fr) 2007-09-04 2009-03-12 Curevac Gmbh Complexes d'arn et de peptides cationiques pour transfection et immunostimulation
WO2009046739A1 (fr) 2007-10-09 2009-04-16 Curevac Gmbh Composition pour traiter le cancer de la prostate (pca)
WO2009046738A1 (fr) 2007-10-09 2009-04-16 Curevac Gmbh Composition pour traiter le cancer du poumon, notamment les cancers du poumon non à petites cellules (nsclc)
EP3346005A1 (fr) 2008-01-31 2018-07-11 CureVac AG Acides nucléiques de formule (i) (nugixmgnnv)a et dérivés associés en tant qu'agent/adjuvant de stimulation immunitaire
WO2010037408A1 (fr) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprenant un arnm complexé et un arnm nu pour déclencher ou augmenter une réponse immunostimulante chez un mammifère et utilisations de ladite composition
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
WO2011069529A1 (fr) 2009-12-09 2011-06-16 Curevac Gmbh Solution contenant du mannose pour la lyophilisation, la transfection et/ou l'injection d'acides nucléiques
AU2011285200B2 (en) 2010-07-30 2014-08-21 CureVac SE Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation
WO2012019630A1 (fr) 2010-08-13 2012-02-16 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'une protéine codée
WO2012089225A1 (fr) 2010-12-29 2012-07-05 Curevac Gmbh Combinaison de vaccination et d'inhibition de la présentation des antigènes restreinte par le cmh de classe i
WO2012116715A1 (fr) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination chez des nouveaux-nés et des enfants en bas âge
WO2012113413A1 (fr) 2011-02-21 2012-08-30 Curevac Gmbh Composition de vaccin comprenant des acides nucléiques immunostimulateurs complexés et des antigènes emballés avec des conjugués de polyéthylèneglycol/peptide à liaison disulfure
EP2623121A1 (fr) 2012-01-31 2013-08-07 Bayer Innovation GmbH Composition pharmaceutique comportant un complexe de chargement de porteur polymérique et un antigène
WO2013113325A1 (fr) 2012-01-31 2013-08-08 Curevac Gmbh Complexes chargés négativement comprenant des acides nucléiques pour l'immunostimulation
WO2013113326A1 (fr) 2012-01-31 2013-08-08 Curevac Gmbh Composition pharmaceutique comprenant un complexe support polymère - charge et au moins un antigène de protéine ou de peptide
WO2013120499A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'un antigène pathogène codé
WO2013120497A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour l'augmentation de l'expression d'une protéine thérapeutique codée
WO2013120500A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation en vue d'augmenter l'expression d'un antigène tumoral codé
WO2013120498A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'un autoantigène auto-immun ou d'un antigène allergène codé
CN104284979B (zh) 2012-03-27 2018-06-08 库瑞瓦格股份公司 用于提高的蛋白或肽表达的人工核酸分子
CN108929880A (zh) 2012-03-27 2018-12-04 库瑞瓦格股份公司 包含5′toputr的人工核酸分子
SG11201405542UA (en) 2012-03-27 2014-10-30 Curevac Gmbh Artificial nucleic acid molecules
ES2719598T3 (es) 2012-05-25 2019-07-11 Curevac Ag Inmovilización reversible y/o liberación controlada de ácidos nucleicos contenidos en nanopartículas mediante revestimientos poliméricos (biodegradables)
SG11201506052PA (en) 2013-02-22 2015-09-29 Curevac Gmbh Combination of vaccination and inhibition of the pd-1 pathway
CN105473158B (zh) 2013-08-21 2021-04-13 库瑞瓦格股份公司 呼吸道合胞病毒(rsv)疫苗
SG10201801428RA (en) 2013-08-21 2018-03-28 Curevac Ag Method for increasing expression of rna-encoded proteins
BR112016000889A2 (pt) 2013-08-21 2017-12-12 Curevac Ag composição e vacina para tratamento de câncer de próstata
HUE046469T2 (hu) 2013-08-21 2020-03-30 Curevac Ag Készítmény és vakcina tüdõrák kezelésére
AU2014310931B2 (en) 2013-08-21 2019-12-19 CureVac SE Rabies vaccine
WO2015024669A1 (fr) 2013-08-21 2015-02-26 Curevac Gmbh Vaccin combiné
ES2806575T3 (es) 2013-11-01 2021-02-18 Curevac Ag ARN modificado con propiedades inmunoestimuladoras disminuidas
SG10201805660WA (en) 2013-12-30 2018-08-30 Curevac Ag Methods for rna analysis
RU2717986C2 (ru) 2013-12-30 2020-03-27 Куревак Аг Искусственные молекулы нуклеиновой кислоты
WO2015101415A1 (fr) 2013-12-30 2015-07-09 Curevac Gmbh Molécules d'acides nucléiques artificielles
WO2015135558A1 (fr) 2014-03-12 2015-09-17 Curevac Gmbh Combinaison de vaccination et d'agonistes de ox40
WO2015149944A2 (fr) 2014-04-01 2015-10-08 Curevac Gmbh Complexe cargo de support polymère à utiliser comme agent immunostimulant ou comme adjuvant

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011008974A2 (fr) * 2009-07-15 2011-01-20 Novartis Ag Compositions à base de protéine f du vrs et procédés de fabrication associés
WO2012116811A1 (fr) * 2011-03-02 2012-09-07 Curevac Gmbh Vaccination chez des patients âgés

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BENJAMIN PETSCH ET AL: "Protective efficacy of in vitro synthesized, specific mRNA vaccines against influenza A virus infection", NATURE BIOTECHNOLOGY, vol. 30, no. 12, 1 January 2012 (2012-01-01), pages 1210 - 1216, XP055051005, ISSN: 1087-0156, DOI: 10.1038/nbt.2436 *
FLEETON M N ET AL: "Self-replicative RNA vaccines elicit protection against influenza A virus, respiratory syncytial virus, and a tickborne encephalitis virus", JOURNAL OF INFECTIOUS DISEASES. JID, UNIVERSITY OF CHICAGO PRESS, CHICAGO, IL, vol. 183, no. 9, 1 May 2001 (2001-05-01), pages 1395 - 1398, XP002224503, ISSN: 0022-1899, DOI: 10.1086/319857 *
S. DE BAETS ET AL: "Recombinant Influenza Virus Carrying the Respiratory Syncytial Virus (RSV) F85-93 CTL Epitope Reduces RSV Replication in Mice", JOURNAL OF VIROLOGY, vol. 87, no. 6, 9 January 2013 (2013-01-09), pages 3314 - 3323, XP055148479, ISSN: 0022-538X, DOI: 10.1128/JVI.03019-12 *
TALAAT A M ET AL: "A combination vaccine confers full protection against co-infections with influenza, herpes simplex and respiratory syncytial viruses", VACCINE, ELSEVIER LTD, GB, vol. 20, no. 3-4, 12 November 2001 (2001-11-12), pages 538 - 544, XP027321869, ISSN: 0264-410X, [retrieved on 20011112] *
TIFFANY M TURNER ET AL: "A novel influenza virus hemagglutinin-respiratory syncytial virus (RSV) fusion protein subunit vaccine against influenza and RSV", JOURNAL OF VIROLOGY, AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 87, no. 19, 31 July 2013 (2013-07-31), pages 10792 - 10804, XP002722825, ISSN: 1098-5514, [retrieved on 20130731], DOI: 10.1128/JVI.01724-13 *

Cited By (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11975064B2 (en) 2011-03-02 2024-05-07 CureVac SE Vaccination with mRNA-coded antigens
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US11965000B2 (en) 2013-08-21 2024-04-23 CureVac SE Respiratory syncytial virus (RSV) vaccine
US11739125B2 (en) 2013-08-21 2023-08-29 Cure Vac SE Respiratory syncytial virus (RSV) vaccine
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US10022435B2 (en) 2014-04-23 2018-07-17 Modernatx, Inc. Nucleic acid vaccines
WO2016011401A1 (fr) * 2014-07-18 2016-01-21 Kedl Ross M Combinaisons d'immunostimulation et leur utilisation
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US11667910B2 (en) 2015-05-29 2023-06-06 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
US11760992B2 (en) 2015-05-29 2023-09-19 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
US11834651B2 (en) 2015-05-29 2023-12-05 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
EP4286012A2 (fr) 2015-09-17 2023-12-06 ModernaTX, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
EP3736261A1 (fr) 2015-09-17 2020-11-11 ModernaTX, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2017049245A2 (fr) 2015-09-17 2017-03-23 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2017066782A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffes d'arnm hydrophobes
EP4086269A1 (fr) 2015-10-16 2022-11-09 ModernaTX, Inc. Analogues de capuchon d'arnm avec liaison de phosphate modifiée
WO2017066789A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm avec sucre modifié
WO2017066781A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm à liaison phosphate modifié
WO2017066791A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffe d'arnm à substitution sucre
WO2017066793A1 (fr) 2015-10-16 2017-04-20 Modernatx, Inc. Analogues de coiffes arnm et procédés de coiffage d'arnm
AU2016342048B2 (en) * 2015-10-22 2022-09-08 Modernatx, Inc. Broad spectrum influenza virus vaccine
WO2017070620A3 (fr) * 2015-10-22 2017-07-13 Modernatx, Inc. Vaccin contre le virus de la grippe à large spectre
US11786590B2 (en) 2015-11-09 2023-10-17 CureVac SE Rotavirus vaccines
EP4036079A2 (fr) 2015-12-22 2022-08-03 ModernaTX, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques et/ou prophylactiques
WO2017112865A1 (fr) 2015-12-22 2017-06-29 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques et/ou prophylactiques
WO2017109134A1 (fr) * 2015-12-22 2017-06-29 Curevac Ag Procédé de production de compositions de molécules d'arn
CN108778308A (zh) * 2015-12-22 2018-11-09 库瑞瓦格股份公司 生产rna分子组合物的方法
EP3701963A1 (fr) 2015-12-22 2020-09-02 CureVac AG Procédé de production de compositions de molécules d'arn
JP2019503678A (ja) * 2015-12-22 2019-02-14 キュアバック アーゲー Rna分子組成物の作製方法
US11684665B2 (en) 2015-12-22 2023-06-27 CureVac SE Method for producing RNA molecule compositions
EP4233898A3 (fr) * 2016-05-04 2023-11-01 CureVac SE Vaccins contre l'arnm de la grippe
WO2017191258A1 (fr) * 2016-05-04 2017-11-09 Curevac Ag Vaccins contre l'arnm de la grippe
WO2017218704A1 (fr) 2016-06-14 2017-12-21 Modernatx, Inc. Formulations stabilisées de nanoparticules lipidiques
WO2018089540A1 (fr) 2016-11-08 2018-05-17 Modernatx, Inc. Formulations stabilisées de nanoparticules lipidiques
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
US11696946B2 (en) 2016-11-11 2023-07-11 Modernatx, Inc. Influenza vaccine
EP4035659A1 (fr) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes destinés à l'administration d'agents thérapeutiques
US11524066B2 (en) 2016-12-23 2022-12-13 CureVac SE Henipavirus vaccine
WO2018115507A3 (fr) * 2016-12-23 2018-08-30 Curevac Ag Vaccin contre l'hénipavirus
US11865084B2 (en) 2016-12-23 2024-01-09 CureVac SE MERS coronavirus vaccine
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
WO2018170306A1 (fr) 2017-03-15 2018-09-20 Modernatx, Inc. Composés et compositions d'administration intracellulaire d'agents thérapeutiques
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
WO2018170336A1 (fr) 2017-03-15 2018-09-20 Modernatx, Inc. Formulation de nanoparticules lipidiques
EP4186888A1 (fr) 2017-03-15 2023-05-31 ModernaTX, Inc. Composé et compositions pour l'administration intracellulaire d'agents thérapeutiques
EP3595676A4 (fr) * 2017-03-17 2021-05-05 Modernatx, Inc. Vaccins à base d'arn contre des maladies zoonotiques
US11497807B2 (en) 2017-03-17 2022-11-15 Modernatx, Inc. Zoonotic disease RNA vaccines
WO2018232120A1 (fr) 2017-06-14 2018-12-20 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents
WO2019036638A1 (fr) 2017-08-18 2019-02-21 Modernatx, Inc. Procédés de préparation d'arn modifié
WO2019046809A1 (fr) 2017-08-31 2019-03-07 Modernatx, Inc. Procédés de fabrication de nanoparticules lipidiques
WO2019148101A1 (fr) * 2018-01-29 2019-08-01 Modernatx, Inc. Vaccins à base d'arn contre le vrs
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines
US11433139B2 (en) 2018-03-16 2022-09-06 Zoetis Services Llc Peptide vaccines against interleukin-31
WO2020061367A1 (fr) 2018-09-19 2020-03-26 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2020061457A1 (fr) 2018-09-20 2020-03-26 Modernatx, Inc. Préparation de nanoparticules lipidiques et leurs méthodes d'administration
WO2020160430A1 (fr) 2019-01-31 2020-08-06 Modernatx, Inc. Mélangeurs à tourbillon et procédés, systèmes, et appareils associés
WO2020160397A1 (fr) 2019-01-31 2020-08-06 Modernatx, Inc. Procédés de préparation de nanoparticules lipidiques
WO2021204175A1 (fr) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Compositions de nanoparticules lipidiques
WO2021204179A1 (fr) 2020-04-09 2021-10-14 Suzhou Abogen Biosciences Co., Ltd. Vaccins à base d'acide nucléique pour coronavirus
WO2022002040A1 (fr) 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2022037652A1 (fr) 2020-08-20 2022-02-24 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2022152141A2 (fr) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques conjugués polymères et compositions de nanoparticules lipidiques
WO2022152109A2 (fr) 2021-01-14 2022-07-21 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2022247755A1 (fr) 2021-05-24 2022-12-01 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2023044333A1 (fr) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Lipides cycliques et leurs procédés d'utilisation
WO2023044343A1 (fr) 2021-09-14 2023-03-23 Renagade Therapeutics Management Inc. Lipides acycliques et leurs procédés d'utilisation
EP4162950A1 (fr) 2021-10-08 2023-04-12 Suzhou Abogen Biosciences Co., Ltd. Vaccins d'acide nucléique pour coronavirus
WO2023056917A1 (fr) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2023056914A1 (fr) 2021-10-08 2023-04-13 Suzhou Abogen Biosciences Co., Ltd. Composés lipidiques et compositions de nanoparticules lipidiques
WO2023122752A1 (fr) 2021-12-23 2023-06-29 Renagade Therapeutics Management Inc. Lipides contraints et procédés d'utilisation associés
WO2023196931A1 (fr) 2022-04-07 2023-10-12 Renagade Therapeutics Management Inc. Lipides cycliques et nanoparticules lipidiques (npl) pour l'apport d'acides nucléiques ou de peptides destinés à être utilisés dans la vaccination contre des agents infectieux
WO2024037578A1 (fr) 2022-08-18 2024-02-22 Suzhou Abogen Biosciences Co., Ltd. Composition de nanoparticules lipidiques
WO2024089634A1 (fr) * 2022-10-27 2024-05-02 Pfizer Inc. Compositions immunogènes contre la grippe et le vrs

Also Published As

Publication number Publication date
US11266735B2 (en) 2022-03-08
US10588959B2 (en) 2020-03-17
AU2014310935A1 (en) 2016-01-21
US20200155668A1 (en) 2020-05-21
US20160166678A1 (en) 2016-06-16
CA2915730A1 (fr) 2015-02-26
US20220152193A1 (en) 2022-05-19
AU2014310935B2 (en) 2019-11-21
CN105473157A (zh) 2016-04-06

Similar Documents

Publication Publication Date Title
US11266735B2 (en) Combination vaccine
US11739125B2 (en) Respiratory syncytial virus (RSV) vaccine
US11110166B2 (en) Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
US20200268908A1 (en) Rabies vaccine
US20210162037A1 (en) Influenza mrna vaccines
EP2680880B2 (fr) Vaccinations chez les nouveau-nés et les bébés
EP3035959A1 (fr) Vaccin combiné
EP3035960B1 (fr) Vaccin contre le virus respiratoire syncytial
EP4043032A1 (fr) Vaccin antirabique

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201480044258.X

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14761282

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2915730

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2014310935

Country of ref document: AU

Date of ref document: 20140821

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2014761282

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE